1
|
Lv X, Yang C, Li X, Liu Y, Yang Y, Jin T, Chen Z, Jia J, Wang M, Li L. Ferroptosis and hearing loss: from molecular mechanisms to therapeutic interventions. J Enzyme Inhib Med Chem 2025; 40:2468853. [PMID: 39992186 PMCID: PMC11852237 DOI: 10.1080/14756366.2025.2468853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 02/11/2025] [Accepted: 02/13/2025] [Indexed: 02/25/2025] Open
Abstract
Hearing loss profoundly affects social engagement, mental health, cognition, and brain development, with sensorineural hearing loss (SNHL) being a major concern. Linked to ototoxic medications, ageing, and noise exposure, SNHL presents significant treatment challenges, highlighting the need for effective prevention and regeneration strategies. Ferroptosis, a distinct form of cell death featuring iron-dependent lipid peroxidation, has garnered interest due to its potential role in cancer, ageing, and neuronal degeneration, especially hearing loss. The emerging role of ferroptosis as a crucial mediator in SNHL suggests that it may offer a novel therapeutic target for otoprotection. This review aims to summarise the intricate connection between ferroptosis and SNHL, offering a fresh perspective for exploring targeted therapeutic strategies that could potentially mitigate cochlear cells damage and enhance the quality of life for individuals with hearing impairments.
Collapse
Affiliation(s)
- Xingyi Lv
- Department of Physiology, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, China
| | - Chenyi Yang
- Department of Physiology, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, China
| | - Xianying Li
- Department of Physiology, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, China
| | - Yun Liu
- Department of Physiology, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, China
| | - Yu Yang
- Department of Physiology, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, China
| | - Tongyan Jin
- Department of Physiology, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, China
| | - Zhijian Chen
- Department of Physiology, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, China
| | - Jinjing Jia
- Department of Physiology, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, China
| | - Min Wang
- Department of Physiology, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, China
| | - Li Li
- Department of Physiology, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, China
| |
Collapse
|
2
|
Xi Y, Huang Y, Hu J, Wang Y, Qian Q, Tu L, Nie H, Zhu J, Ding C, Gao X, Zheng X, Huang D, Cheng L. EIF2B5 promotes malignant progression of hepatocellular carcinoma by activating the PI3K/AKT signaling pathway through targeting RPL6. Cell Signal 2025; 132:111821. [PMID: 40246131 DOI: 10.1016/j.cellsig.2025.111821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 04/05/2025] [Accepted: 04/15/2025] [Indexed: 04/19/2025]
Abstract
Hepatocellular carcinoma (HCC) is a highly aggressive malignancy with limited treatment options and poor prognosis. In this study, we demonstrated the critical role of EIF2B5 in driving HCC progression. We found EIF2B5 expression is significantly upregulated in HCC tumor tissues in several bioinformatics datasets, including The Cancer Genome Atlas, and that high expression of EIF2B5 predicts poor prognosis for HCC patients. Through a series of in vitro cell biology experiments, we found that EIF2B5 knockdown significantly attenuated Hep3B and HepG2 proliferation, migration, and invasion and increased cell cycle arrest, whereas EIF2B5 overexpression promoted HCC progression. Through mass spectrometry and immunoprecipitation validation, we found that EIF2B5 directly interacted with RPL6 and that when EIF2B5 was overexpressed in HCC cells, it promoted the expression of the downstream protein RPL6, which was able to activate the phosphatidylinositol kinase (PI3K)/serine-threonine kinase (AKT)/mammalian target of rapamycin (mTOR) pathway and thereby increase the proliferation and invasion ability of HCC cell lines, as verified by second-generation sequencing analysis and western blot. We further verified these findings using the mouse ectopic tumor assay, and the results showed that EIF2B5 knockdown significantly inhibited tumor progression in HCC mice. The present study suggests that EIF2B5 promotes malignant progression of HCC by interacting with RPL6 and activating the PI3K/AKT/mTOR signaling pathway and may serve as a potential target for the treatment of HCC.
Collapse
Affiliation(s)
- Yiling Xi
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yue Huang
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Jiahui Hu
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yan Wang
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Qiyi Qian
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Linglan Tu
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Huizong Nie
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Jiayao Zhu
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Chenguang Ding
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Xiaotao Gao
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Xiaoliang Zheng
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Dongsheng Huang
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Liyan Cheng
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang, China.
| |
Collapse
|
3
|
Gao YF, Yang YJ, Qin JB, Yu MY, Hu SW, Zhang HF, Lin FH, Hu HY, Fang MJ, Zeng JZ. Design, synthesis, and biological evaluation of quinolinyl-ureido-phenyl-hydrazide derivatives and quinolinyl-hydrazide derivatives as anticancer agents targeting Nur77-mediated ferroptosis. Eur J Med Chem 2025; 291:117559. [PMID: 40215561 DOI: 10.1016/j.ejmech.2025.117559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Revised: 03/11/2025] [Accepted: 03/21/2025] [Indexed: 05/03/2025]
Abstract
In the recent decade, targeting ferroptosis for cancer therapy has attracted remarkable attention. Interestingly, the transcriptional regulator Nur77, a promising therapeutic target in cancer, has been recently identified as a crucial regulator of ferroptosis. However, no ferroptosis inducer targeting Nur77 has been reported currently. In this study, we built upon our prior research on Nur77 modulator 4-PQBH to design and synthesize four series of new compounds, with the objective of developing novel Nur77-mediated ferroptosis inducers. Among them, compound 8f exhibited the most potency against the tested cancer cell lines, including human estrogen positive breast cancer and triple-negative breast cancer cell lines, while displaying lower toxicity towards human normal cell lines HaCaT and MCF-10A (IC50> 50 μM). Furthermore, 8f demonstrated superior Nur77-binding activity in comparison to the reference compound Csn-B, and it has the capacity to activate the Nur77-driven luciferase activity and increase the protein level of Nur77. Remarkably, 8f induced an increase in the levels of reactive oxygen species (ROS), malondialdehyde (MDA), and lipid peroxidation, concurrently with a reduction in the expression of GPX4 protein, culminating in the induction of ferroptosis in a Nur77-dependent manner. In vivo, 8f treatment has been observed to significantly suppress MCF7 xenograft tumor growth. Consequently, a novel ferroptosis inducer targeting Nur77 (8f) is first reported as a potent anti-EPBC agent, providing may serve as a promising lead for further drug development targeting Nur77-mediated ferroptosis.
Collapse
Affiliation(s)
- Yan-Fang Gao
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, 361102, China
| | - Yi-Jing Yang
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, 361102, China
| | - Jing-Bo Qin
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, 361102, China; Guangdong Provincial Engineering Research Center of Molecular Imaging, Guangdong-Hong Kong-Macao University Joint Laboratory of the Interventional Medicine Foundation of Guangdong Province, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, 519000, China
| | - Ming-Yue Yu
- Xingzhi College, Zhejiang Normal University, Lanxi, 321004, China; College of Chemistry and Bioengineering, Yichun, 336000, China
| | - Sheng-Wei Hu
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, 361102, China
| | - Hao-Fan Zhang
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, 361102, China
| | - Fan-Hong Lin
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, 361102, China
| | - Hong-Yu Hu
- Xingzhi College, Zhejiang Normal University, Lanxi, 321004, China.
| | - Mei-Juan Fang
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, 361102, China.
| | - Jin-Zhang Zeng
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, 361102, China.
| |
Collapse
|
4
|
Shen K, Hu C, Zhang Y, Cheng X, Xu Z, Pan S. Advances and applications of multiomics technologies in precision diagnosis and treatment for gastric cancer. Biochim Biophys Acta Rev Cancer 2025; 1880:189336. [PMID: 40311712 DOI: 10.1016/j.bbcan.2025.189336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Revised: 04/24/2025] [Accepted: 04/25/2025] [Indexed: 05/03/2025]
Abstract
Gastric cancer (GC), one of the most prevalent malignancies worldwide, is distinguished by extensive genetic and phenotypic heterogeneity, posing persistent challenges to conventional diagnostic and therapeutic strategies. The significant global burden of GC highlights an urgent need to unravel its complex underlying mechanisms, discover novel diagnostic and prognostic biomarkers, and develop more effective therapeutic interventions. In this context, this review comprehensively examines the transformative roles of cutting-edge technologies, including radiomics, pathomics, genomics, transcriptomics, epigenomics, proteomics, and metabolomics, in advancing precision diagnosis and treatment for GC. Multiomics data analysis not only deepens our understanding of GC pathogenesis and molecular subtypes but also identifies promising biomarkers, facilitating the creation of tailored therapeutic approaches. Additionally, integrating multiomics approaches holds immense potential for elucidating drug resistance mechanisms, predicting patient outcomes, and uncovering novel therapeutic targets, thereby laying a robust foundation for precision medicine in the comprehensive management of GC.
Collapse
Affiliation(s)
- Ke Shen
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China; Postgraduate Training Base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, China
| | - Can Hu
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China; Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, Zhejiang 310022, China; Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, 310022, China
| | - Yanqiang Zhang
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China; Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, Zhejiang 310022, China; Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, 310022, China
| | - Xiangdong Cheng
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China; Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, Zhejiang 310022, China; Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, 310022, China
| | - Zhiyuan Xu
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China; Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, Zhejiang 310022, China; Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, 310022, China.
| | - Siwei Pan
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China; Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, Zhejiang 310022, China; Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, 310022, China.
| |
Collapse
|
5
|
Schnabl SD, Klubien J, O'Rourke CJ, Bull Nordkild S, Kugler JM, Dam Nielsen S, Andersen JB, Pommergaard HC. Validation of Two Prognostic Gene Scores in Patients Undergoing Liver Resection for Hepatocellular Carcinoma. J Clin Exp Hepatol 2025; 15:102544. [PMID: 40248345 PMCID: PMC12002650 DOI: 10.1016/j.jceh.2025.102544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 03/04/2025] [Indexed: 04/19/2025] Open
Abstract
Background/Aims Several prognostic gene signatures have been proposed as predictors of the prognosis of hepatocellular carcinoma (HCC), yet none are implemented in the clinical setting. We aimed to validate two gene scores previously derived from European cohorts. Methods The patients who underwent liver resection for HCC at Copenhagen University Hospital, Rigshospitalet from 2014 to 2018 were included. RNA sequencing determined the expression of genes in the '5-gene score' (HN1, RAN, RAMP3, KRT19, TAF9B) and 'HepatoPredict' (CLU, DPT, SPRY2, CAPSN1). Univariable Cox regression assessed associations with overall and disease-free survival. These parameters were also analyzed in the The Cancer Genome Atlas Liver Hepatocellular Carcinoma (TCGA-LIHC) (n = 359) and National Institute of Health (NIH) (n = 178) cohorts. Results Among 51 patients (88% male), 59% had no underlying liver disease and 25% had cirrhosis. No individual genes were significantly associated with overall survival in the Danish cohort. In the TCGA-LIHC cohort, CLU was linked to better overall survival, and in the NIH cohort, high expression of SPRY2 was associated with poorer overall survival. In the TCGA-LIHC cohort, HN1, RAN, and TAF9B were associated with poorer overall survival, while RAMP3 was linked to better overall survival. No genes were associated with disease-free survival. Conclusion Few individual genes significantly predicted survival in the larger cohorts, and none in the Danish cohort. However, the clinical implication of this needs further investigation.
Collapse
Affiliation(s)
- Stinna D. Schnabl
- Department of Surgery and Transplantation, Copenhagen University Hospital, Rigshospitalet, Denmark
- Hepatic Malignancy Surgical Research Unit (HEPSURU), Department of Surgery and Transplantation, Rigshospitalet, Copenhagen University Hospital, Denmark
| | - Jeanett Klubien
- Department of Surgery and Transplantation, Copenhagen University Hospital, Rigshospitalet, Denmark
- Hepatic Malignancy Surgical Research Unit (HEPSURU), Department of Surgery and Transplantation, Rigshospitalet, Copenhagen University Hospital, Denmark
| | - Colm J. O'Rourke
- Biotech Research and Innovation Centre (BRIC), Department of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Sophie Bull Nordkild
- Department of Surgery and Transplantation, Copenhagen University Hospital, Rigshospitalet, Denmark
- Hepatic Malignancy Surgical Research Unit (HEPSURU), Department of Surgery and Transplantation, Rigshospitalet, Copenhagen University Hospital, Denmark
| | - Jan-Michael Kugler
- Institute for Molecular and Cellular Medicine, University of Copenhagen, Panum Institute, Copenhagen, Denmark
| | - Susanne Dam Nielsen
- Department of Surgery and Transplantation, Copenhagen University Hospital, Rigshospitalet, Denmark
- Viro-immunology Research Unit, Department of Infectious Diseases, Copenhagen University Hospital, Rigshospitalet, Denmark
- Institute for Clinical Medicine, University of Copenhagen, Panum Institute, Copenhagen, Denmark
| | - Jesper B. Andersen
- Biotech Research and Innovation Centre (BRIC), Department of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Hans-Christian Pommergaard
- Department of Surgery and Transplantation, Copenhagen University Hospital, Rigshospitalet, Denmark
- Hepatic Malignancy Surgical Research Unit (HEPSURU), Department of Surgery and Transplantation, Rigshospitalet, Copenhagen University Hospital, Denmark
- Institute for Clinical Medicine, University of Copenhagen, Panum Institute, Copenhagen, Denmark
| |
Collapse
|
6
|
Qiu J, Guo F, Shi J, Guo T, Piao H. Piperlongumine inhibits glioblastoma proliferation by inducing ferroptosis. J Pharm Pharmacol 2025; 77:822-833. [PMID: 39692457 DOI: 10.1093/jpp/rgae148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 11/06/2024] [Indexed: 12/19/2024]
Abstract
OBJECTIVES This study aimed to investigate the effects of Piperlongumine on Glioblastoma multiforme. METHODS The effects of Piperlongumine on the viability and proliferation of glioma cells LN229 and A172 were measured. Changes in mitochondrial structure were observed. Cell proliferative capacity was assessed using immunofluorescence. The levels of glutathione, malondialdehyde, 4-hydroxynonenal, and intracellular reactive oxygen species were detected. The levels of ferroptosis-related proteins were detected. A plasmid transfection was performed to overexpress the nuclear factor erythroid 2-related factor 2 gene; a subcutaneous tumor model was established in nude mice to observe the in vivo inhibitory effects of Piperlongumine on Glioblastoma multiforme and the recovery effect of Fer-1. The expression levels of ferroptosis-related proteins were detected using immunohistochemistry. KEY FINDINGS Piperlongumine inhibited the viability of glioma cells, as well as their proliferation. The ferroptosis inhibitors were able to restore the inhibitory effect of Piperlongumine on glioma cell proliferation. Forced overexpression of nuclear factor erythroid 2-related factor 2 partially reversed Piperlongumine-induced ferroptosis; Piperlongumine exhibited a significant inhibitory effect on Glioblastoma multiforme cells in vivo, which could be restored by Fer-1. CONCLUSIONS Piperlongumine inhibits Glioblastoma multiforme proliferation by inducing ferroptosis in vitro and vivo model.
Collapse
Affiliation(s)
- Jianting Qiu
- Liaoning University of Traditional Chinese Medicine, Shenyang 110000, China
- Department of Neurology, The People's Hospital of Liaoning Province, Shenyang 110000, China
| | - Fangzhou Guo
- Department of Neurosurgery, Guangxi Medical University Cancer Hospital & Guangxi Cancer Institute, Nanning 530000, China
| | - Ji Shi
- Department of Neurosurgery, Liaoning Cancer Hospital & Institute, Shenyang 110000, China
| | - Tangjun Guo
- Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221000, China
| | - Haozhe Piao
- Liaoning University of Traditional Chinese Medicine, Shenyang 110000, China
- Department of Neurosurgery, Liaoning Cancer Hospital & Institute, Shenyang 110000, China
| |
Collapse
|
7
|
Wang D, Li L, Zhang F, Liu Q, Wang WL, Wang ZX, Wu YT, Bu PL. SLC1A5 mediates myocardial damage after myocardial infarction by triggering cardiomyocyte ferroptosis. Biochem Pharmacol 2025:117047. [PMID: 40490039 DOI: 10.1016/j.bcp.2025.117047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2025] [Revised: 05/05/2025] [Accepted: 06/05/2025] [Indexed: 06/11/2025]
Abstract
Myocardial infarction (MI) has become a major disease that causes significant global mortality. Notably ferroptosis may exert a key effect on myocardial injury after MI. As the glutamine transporter on the cell membrane, solute carrier family 1 member 5 (SLC1A5) plays a role as a ferroptosis-inducing gene and has a mediating effect on cell ferroptosis. However, whether SLC1A5 is involved in mediating cardiomyocyte ferroptosis and myocardial injury after MI remains to be further elucidated. In the present study, we investigated whether SLC1A5 mediated myocardial injury after MI by triggering cardiomyocyte ferroptosis in vivo and in vitro. Based on our findings, SLC1A5 exhibited crucial mediating effects on post-MI cardiomyocyte ferroptosis and myocardial injury, and these effects were stimulated by SLC1A5 overexpression, but inhibited by ferrostatin-1 (a ferroptosis inhibitor)and V9302 (a SLC1A5 inhibitor). In conclusion, our results revealed a novel molecular mechanism of ferroptosis regulated by SLC1A5, which is essential for cardiomyocyte ferroptosis pathogenesis and myocardial injury post-MI.
Collapse
Affiliation(s)
- Dong Wang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan 250012, China; Binzhou Medical University Hospital, Binzhou 256603, China
| | - Lei Li
- Binzhou Medical University Hospital, Binzhou 256603, China
| | - Feng Zhang
- Binzhou Medical University Hospital, Binzhou 256603, China
| | - Qian Liu
- Binzhou Medical University Hospital, Binzhou 256603, China
| | - Wen-Long Wang
- Binzhou Medical University Hospital, Binzhou 256603, China
| | - Zhi-Xin Wang
- Binzhou Medical University Hospital, Binzhou 256603, China
| | - Yu-Ting Wu
- Binzhou Medical University Hospital, Binzhou 256603, China; School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China.
| | - Pei-Li Bu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan 250012, China.
| |
Collapse
|
8
|
Wang R, Dai F, Deng Z, Tang L, Liu H, Xia L, Cheng Y. ITGA3 participates in the pathogenesis of recurrent spontaneous abortion by downregulating ULK1-mediated autophagy to inhibiting trophoblast function. Am J Physiol Cell Physiol 2025; 328:C1941-C1956. [PMID: 39437445 DOI: 10.1152/ajpcell.00563.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 09/04/2024] [Accepted: 10/12/2024] [Indexed: 10/25/2024]
Abstract
Recurrent spontaneous abortion (RSA) is a significant challenge encountered by couples of reproductive ages, with inadequate trophoblast invasion identified as a primary factor in RSA pathogenesis. However, the precise molecular mechanisms through which trophoblast cell dysfunction leads to RSA remain incompletely understood. Research has highlighted the critical role of integrins in embryo implantation and development. Although integrin α-3 (ITGA3) is recognized for its promotion of invasion in cancer cells, its involvement in miscarriage remains poorly characterized. This investigation initially assessed ITGA3 expression in villous tissues obtained from patients with RSA and patients with induced abortion. The findings demonstrated a notable reduction in ITGA3 levels in the villous tissues of patients with RSA compared with the control group. Subsequent in vitro analyses indicated that ITGA3 knockdown inhibited the migration, invasion, and proliferation of trophoblast cells. Through RNA sequencing and subsequent experimentation, it was revealed that ITGA3 regulated Unc51-like kinase 1 (ULK1)-mediated autophagy to influence trophoblast cell invasion, migration, and proliferation. Furthermore, utilizing a miscarriage animal model, the diminished expression of ITGA3 and ULK1 in the placentas of RSA mice was confirmed. In conclusion, the study findings suggest that the downregulation of ITGA3 suppresses ULK1 expression, consequently impeding autophagy to initiation and impeding trophoblast cell invasion and migration, thereby contributing to the pathological progression of RSA.NEW & NOTEWORTHY There is a strong correlation between the reduced expression of ITGA3 in villous tissues and RSA. ITGA3 facilitates the expression of ULK1, thereby promoting autophagy formation and elevating autophagy levels in trophoblast cells. Consequently, this enhances the invasion and migration abilities of trophoblast cells.
Collapse
Affiliation(s)
- Ruiqi Wang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
| | - Fangfang Dai
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
| | - Zhimin Deng
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
| | - Lujia Tang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
| | - Hua Liu
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
| | - Liangbin Xia
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
| | - Yanxiang Cheng
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
| |
Collapse
|
9
|
Cheng C, Lin S, Zhu A, Hong Z, Shi Z, Deng H, Zhang G. Linc00239 Facilitates the Progress of Clear Cell Renal Cell Carcinoma via the miR-204-5p/RAB22A Axis. Mol Biotechnol 2025; 67:2357-2366. [PMID: 38850457 PMCID: PMC12055932 DOI: 10.1007/s12033-024-01202-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 11/15/2023] [Indexed: 06/10/2024]
Abstract
Long intergenic non-coding RNA 239 (Linc00239) acts as an oncogene in colorectal cancer (CRC), esophageal squamous cell carcinoma, and acute myeloid leukemia cells. However, its role and regulatory mechanisms in clear cell renal cell carcinoma (ccRCC) remain unknown. We used StarBase and The Cancer Genome Atlas databases to evaluate Linc00239 expression and its effect on ccRCC. Furthermore, the function of Linc00239 in ccRCC proliferation and metastasis was analyzed using Cell Counting Kit-8 and Transwell assays following Linc00239 knockdown. Subsequently, the Linc00239-miRNA-mRNA regulatory associations were selected based on miRanda, miTarbase, and previous references, and their expression levels and binding relationship were further validated using quantitative real-time polymerase chain reaction, western blotting and dual-luciferase reporter gene assay. Additionally, we transfected a miRNA inhibitor to evaluate whether the miR-204-5p/RAB22A (Ras-related proteins in brain 22a) axis was involved in Linc00239 function. Linc00239 was elevated in ccRCC and correlated with poor prognosis. Linc00239 knockdown inhibited ccRCC progression. Additionally, Linc00239 inhibition elevated miR-204-5p expression and repressed RAB22A levels. Moreover, miR-204-5p inhibitors attenuated this inhibitory effect on proliferation, migration, invasion, and RAB22A level when Linc00239 was knocked down. Linc00239 promotes ccRCC proliferation and metastasis by elevating RAB22A expression through the adsorption of miR-204-5p, which provides a clue for the diagnosis and treatment of ccRCC.
Collapse
Affiliation(s)
- Cheng Cheng
- Department of Urology, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Nanchang, 330006, Jiangxi, China
| | - Shuangquan Lin
- Department of Urology, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Nanchang, 330006, Jiangxi, China
| | - Anyi Zhu
- Department of Urology, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Nanchang, 330006, Jiangxi, China.
| | - Zhengdong Hong
- Department of Urology, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Nanchang, 330006, Jiangxi, China
| | - Zimin Shi
- Department of Urology, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Nanchang, 330006, Jiangxi, China
| | - Huanhuan Deng
- Department of Urology, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Nanchang, 330006, Jiangxi, China
| | - Gan Zhang
- Department of Urology, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Nanchang, 330006, Jiangxi, China
| |
Collapse
|
10
|
Alarcón-Veleiro C, López-Calvo I, Berjawi L, Lucio-Gallego S, Mato-Basalo R, Quindos-Varela M, Lesta-Mellid R, Santamarina-Caínzos I, Varela-Rodríguez S, Fraga M, Quintela M, Vizoso-Vázquez A, Arufe MC, Fafián-Labora J. Ferroptosis: An emerging strategy for managing epithelial ovarian cancer. Biomed Pharmacother 2025; 187:118065. [PMID: 40306179 DOI: 10.1016/j.biopha.2025.118065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 03/30/2025] [Accepted: 04/17/2025] [Indexed: 05/02/2025] Open
Abstract
Ferroptosis is a regulated form of cell death characterised by iron-dependent lipid peroxidation, a process intricately linked to cellular redox homeostasis. This form of cell death is induced by the accumulation of intracellular iron and the subsequent generation of reactive oxygen species (ROS), which leads to lipid peroxidation and ultimately cell death. Ferroptosis is distinct from traditional forms of cell death, such as apoptosis, and holds significant therapeutic potential, particularly in cancers harboring rat sarcoma virus (RAS) mutations, such as epithelial ovarian cancer (EOC). EOC is notoriously resistant to conventional therapies and is associated with a poor prognosis. In this review, we examine recent progress in the understanding of ferroptosis, with a particular focus on its redox biology and the complex regulatory networks involved. We also propose a novel classification system for ferroptosis modulators, grouping them into six categories (I, II, III, IV, V and VI) based on their mechanisms of action and their roles in modulating cellular redox status. By refining these categories, we aim to provide deeper insights into the role of ferroptosis in cancer biology, especially in EOC, and to identify potential therapeutic avenues. We propose that further investigation of ferroptosis in the context of redox biology could reveal novel biomarkers and therapeutic targets, offering promising strategies to overcome resistance mechanisms and improve clinical outcomes for patients with EOC and other treatment-resistant cancers.
Collapse
Affiliation(s)
- C Alarcón-Veleiro
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Ciencias de la Salud, INIBIC-Complejo Hospitalario Universitario A Coruña (CHUAC), Centro Interdisciplinar de Química y Biología (CICA), Universidade da Coruña (UDC), A Coruña 15008, Spain
| | - I López-Calvo
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Ciencias de la Salud, INIBIC-Complejo Hospitalario Universitario A Coruña (CHUAC), Centro Interdisciplinar de Química y Biología (CICA), Universidade da Coruña (UDC), A Coruña 15008, Spain; Grupo EXPRELA, Departamento de Bioloxía, Facultade de Ciencias, Rúa da Fraga, A Coruña 15071, Spain; Centro Interdisciplinar de Química de Química y Biología (CICA), Universidade da Coruña (UDC), A Coruña 15008, Spain; Instituto de Investigación Biomédica de A Coruña (INIBIC), Rúa as Xubias 84, A Coruña 15006, Spain
| | - L Berjawi
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Ciencias de la Salud, INIBIC-Complejo Hospitalario Universitario A Coruña (CHUAC), Centro Interdisciplinar de Química y Biología (CICA), Universidade da Coruña (UDC), A Coruña 15008, Spain
| | - S Lucio-Gallego
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Ciencias de la Salud, INIBIC-Complejo Hospitalario Universitario A Coruña (CHUAC), Centro Interdisciplinar de Química y Biología (CICA), Universidade da Coruña (UDC), A Coruña 15008, Spain
| | - R Mato-Basalo
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Ciencias de la Salud, INIBIC-Complejo Hospitalario Universitario A Coruña (CHUAC), Centro Interdisciplinar de Química y Biología (CICA), Universidade da Coruña (UDC), A Coruña 15008, Spain
| | - M Quindos-Varela
- Translational Cancer Research Group, A Coruña Biomedical Research Institute (INIBIC), Carretera del Pasaje s/n, A Coruña 15006, UK; Complexo Hospitalario Universitario de A Coruña (CHUAC), Spain
| | - R Lesta-Mellid
- Translational Cancer Research Group, A Coruña Biomedical Research Institute (INIBIC), Carretera del Pasaje s/n, A Coruña 15006, UK; Complexo Hospitalario Universitario de A Coruña (CHUAC), Spain
| | - I Santamarina-Caínzos
- Translational Cancer Research Group, A Coruña Biomedical Research Institute (INIBIC), Carretera del Pasaje s/n, A Coruña 15006, UK; Complexo Hospitalario Universitario de A Coruña (CHUAC), Spain
| | - S Varela-Rodríguez
- Translational Cancer Research Group, A Coruña Biomedical Research Institute (INIBIC), Carretera del Pasaje s/n, A Coruña 15006, UK; Complexo Hospitalario Universitario de A Coruña (CHUAC), Spain
| | - M Fraga
- Department of Anatomical Pathology, University Hospital Complex A Coruña, As Xubias 84, A Coruña 15006, Spain
| | - M Quintela
- European Cancer Stem Cell Research Institute, Cardiff University, Cardiff CF24 4HQ, UK
| | - A Vizoso-Vázquez
- Grupo EXPRELA, Departamento de Bioloxía, Facultade de Ciencias, Rúa da Fraga, A Coruña 15071, Spain
| | - M C Arufe
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Ciencias de la Salud, INIBIC-Complejo Hospitalario Universitario A Coruña (CHUAC), Centro Interdisciplinar de Química y Biología (CICA), Universidade da Coruña (UDC), A Coruña 15008, Spain.
| | - J Fafián-Labora
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Ciencias de la Salud, INIBIC-Complejo Hospitalario Universitario A Coruña (CHUAC), Centro Interdisciplinar de Química y Biología (CICA), Universidade da Coruña (UDC), A Coruña 15008, Spain.
| |
Collapse
|
11
|
Elimam H, Zaki MB, Abd-Elmawla MA, Darwish HA, Hatawsh A, Aborehab NM, Mageed SSA, Moussa R, Mohammed OA, Abdel-Reheim MA, Doghish AS. Natural products and long non-coding RNAs in prostate cancer: insights into etiology and treatment resistance. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:6349-6368. [PMID: 39825964 DOI: 10.1007/s00210-024-03736-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Accepted: 12/14/2024] [Indexed: 01/20/2025]
Abstract
Globally, the incidence and death rates associated with cancer persist in rising, despite considerable advancements in cancer therapy. Although some malignancies are manageable by a mix of chemotherapy, surgery, radiation, and targeted therapy, most malignant tumors either exhibit poor responsiveness to early identification or endure post-treatment survival. The prognosis for prostate cancer (PCa) is unfavorable since it is a perilous and lethal malignancy. The capacity of phytochemical and nutraceutical chemicals to repress oncogenic lncRNAs and activate tumor suppressor lncRNAs has garnered significant attention as a possible strategy to diminish the development, proliferation, metastasis, and invasion of cancer cells. A potential technique to treat cancer and enhance the sensitivity of cancer cells to existing conventional therapies is the use of phytochemicals with anticancer characteristics. Functional studies indicate that lncRNAs modulate drug resistance, stemness, invasion, metastasis, angiogenesis, and proliferation via interactions with tumor suppressors and oncoproteins. Among them, numerous lncRNAs, such as HOTAIR, PlncRNA1, GAS5, MEG3, LincRNA-21, and POTEF-AS1, support the development of PCa through many molecular mechanisms, including modulation of tumor suppressors and regulation of various signal pathways like PI3K/Akt, Bax/Caspase 3, P53, MAPK cascade, and TGF-β1. Other lncRNAs, in particular, MALAT-1, CCAT2, DANCR, LncRNA-ATB, PlncRNA1, LincRNA-21, POTEF-AS1, ZEB1-AS1, SChLAP1, and H19, are key players in regulating the aforementioned processes. Natural substances have shown promising anticancer benefits against PCa by altering essential signaling pathways. The overexpression of some lncRNAs is associated with advanced TNM stage, metastasis, chemoresistance, and reduced survival. LncRNAs possess crucial clinical and transitional implications in PCa, as diagnostic and prognostic biomarkers, as well as medicinal targets. To impede the progression of PCa, it is beneficial to target aberrant long non-coding RNAs using antisense oligonucleotides or small interfering RNAs (siRNAs). This prevents them from transmitting harmful messages. In summary, several precision medicine approaches may be used to rectify dysfunctional lncRNA regulatory circuits, so improving early PCa detection and eventually facilitating the conquest of this lethal disease. Due to their presence in biological fluids and tissues, they may serve as novel biomarkers. Enhancing PCa treatments mitigates resistance to chemotherapy and radiation.
Collapse
Affiliation(s)
- Hanan Elimam
- Department of Biochemistry, Faculty of Pharmacy, University of Sadat City, Sadat City, 32897, Egypt.
| | - Mohamed Bakr Zaki
- Department of Biochemistry, Faculty of Pharmacy, University of Sadat City, Sadat City, 32897, Egypt
| | - Mai A Abd-Elmawla
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Hebatallah A Darwish
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
- Pharmacology, Toxicology and Biochemistry Department, Faculty of Pharmacy, Future University in Egypt, Cairo, Egypt
| | - Abdulrahman Hatawsh
- Biotechnology School, Nile University, 26Th of July Corridor, Sheikh Zayed City, 12588, Giza, Egypt
| | - Nora M Aborehab
- Department of Biochemistry, Faculty of Pharmacy, Ahram Canadian University, Giza, Egypt
| | - Sherif S Abdel Mageed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, 11829, Cairo, Egypt
| | - Rewan Moussa
- School Faculty of Medicine, Helwan University, Cairo, 11795, Egypt
| | - Osama A Mohammed
- Department of Pharmacology, College of Medicine, University of Bisha, 61922, Bisha, Saudi Arabia
| | | | - Ahmed S Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, , 11829, Cairo, Egypt
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, 11231, Cairo, Egypt
| |
Collapse
|
12
|
Zhou J, Wang J, Yang L, Fu T, Li H, Shan Y, Gao H, Xie C, Zhang L, Zhang M, Ma J, Liu L, Fang H, Jiang D, Xu M, Pan Q, Gu S. N6-methyadenosine-modified YWHAE mRNA promotes proliferation and inhibits ferroptosis in hepatoblastoma by mediating SLC7A11 expression. Oncogene 2025; 44:1634-1645. [PMID: 40074884 DOI: 10.1038/s41388-025-03334-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 02/03/2025] [Accepted: 02/26/2025] [Indexed: 03/14/2025]
Abstract
Hepatoblastoma (HB) is a rare but predominant liver cancer in children, with few treatment choices in advanced stages. YWHAE is closely related to several human diseases and acts as a molecular scaffold for malignant transformation. However, whether YWHAE promotes HB development remains unknown. Conducting RNA and m6A sequencing on HB tissues, we found that YWHAE was upregulated and modified by N6-methyadenosine. Functionally, YWHAE promoted proliferation and inhibited cell death in HB by in vitro and in vivo studies. Mechanistically, METTL3-dependent m6A modification activated YWHAE mRNA expression, and the m6A reader IGF2BP2 recognized and bound to the m6A site on YWHAE mRNA, thereby enhancing the mRNA stability of YWHAE. Interestingly, RNA sequencing revealed that YWHAE knockdown was involved in regulating ferroptosis of HB cells by mediating SLC7A11 expression. Moreover, knockdown of YWHAE significantly increased the levels of lipid ROS and peroxides in HB cells, promoting the susceptibility of HB cells to ferroptosis. In summary, these findings illuminated the role of YWHAE in HB progression and uncovered its relevance to ferroptosis as a new therapeutic target for HB.
Collapse
Affiliation(s)
- Jiquan Zhou
- Department of General Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai, 200127, China
| | - Jing Wang
- Department of General Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai, 200127, China
| | - Liyuan Yang
- Department of General Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai, 200127, China
| | - Tingyi Fu
- Department of General Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai, 200127, China
| | - Hui Li
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai, 200127, China
| | - Yuhua Shan
- Department of General Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai, 200127, China
| | - Hongxiang Gao
- Department of General Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai, 200127, China
| | - Chenjie Xie
- Department of General Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai, 200127, China
| | - Lei Zhang
- Department of General Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai, 200127, China
| | - Min Zhang
- Pediatric Translational Medicine Institute and Pediatric Congenital Heart Disease Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai, 200127, China
| | - Ji Ma
- Department of Laboratory Medicine, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai, 200127, China
| | - Li Liu
- Department of Laboratory Medicine, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai, 200127, China
| | - Houshun Fang
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai, 200127, China
| | - Dapeng Jiang
- Department of General Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai, 200127, China
| | - Min Xu
- Department of General Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai, 200127, China
| | - Qiuhui Pan
- Department of Laboratory Medicine, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai, 200127, China.
| | - Song Gu
- Department of General Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai, 200127, China.
- Department of International Medical, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai, 200127, China.
| |
Collapse
|
13
|
Li X, Zhao Y, Liu Y, Zhang CS, Xu ZY, Wang SR, Zhang TM, Zhang Y, Liang SX, Yan YB. Panx3 mediates ferroptosis via the AKT/mTOR signaling pathway in oral squamous cell carcinoma. Cell Signal 2025; 134:111908. [PMID: 40441467 DOI: 10.1016/j.cellsig.2025.111908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 04/17/2025] [Accepted: 05/26/2025] [Indexed: 06/02/2025]
Abstract
Pannexin 3 (Panx3) has been the subject of numerous studies across different cell types, predominantly concentrating on bone remodeling, wound repair, and dermal inflammation. However, the link between Panx3 and cancer remains minimally explored. Here, we showed for the first time that the expression of Panx3 was lower in oral squamous cell carcinoma (OSCC) tissues than in normal oral mucosa tissues, and was associated with the differentiation of OSCC. We found that overexpression of Panx3 significantly promotes ferroptosis while inhibiting proliferation, migration, and invasion in SCC15 and CAL27 cells. Furthermore, in Panx3-overexpressing OSCC cells, the expression levels of P-AKT, P-mTOR, GPX4, and SLC7A11 were significantly decreased, whereas ACSL4 expression was markedly upregulated. Subsequently, the enhanced ferroptosis was significantly rescued upon the addition of the AKT activator SC79 to Panx3-overexpressing SCC15 and CAL27 cells. The tumor-suppressive role of Panx3 through ferroptosis induction was further confirmed by xenograft assays, which demonstrated significantly inhibited tumor growth. In conclusion, the results suggested that Panx3 overexpression reduced the survival of OSCC cells and inhibited the progression of OSCC by promoting ferroptosis via the inhibition of AKT/mTOR signaling pathway.
Collapse
Affiliation(s)
- Xuan Li
- Tianjin Medical University, 22 Qi-xiang-tai Road, Heping District, Tianjin 300070, China; Department of Oromaxillofacial-Head and Neck Surgery, Tianjin Stomatological Hospital, School of Medicine, Nankai University, 75 Dagu Road, Heping District, Tianjin 300041, China; Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, 75 Dagu Road, Heping District, Tianjin 300041, China
| | - Yan Zhao
- Tianjin Medical University, 22 Qi-xiang-tai Road, Heping District, Tianjin 300070, China; Department of Oromaxillofacial-Head and Neck Surgery, Tianjin Stomatological Hospital, School of Medicine, Nankai University, 75 Dagu Road, Heping District, Tianjin 300041, China; Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, 75 Dagu Road, Heping District, Tianjin 300041, China
| | - Yang Liu
- Tianjin Medical University, 22 Qi-xiang-tai Road, Heping District, Tianjin 300070, China; Department of Oromaxillofacial-Head and Neck Surgery, Tianjin Stomatological Hospital, School of Medicine, Nankai University, 75 Dagu Road, Heping District, Tianjin 300041, China; Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, 75 Dagu Road, Heping District, Tianjin 300041, China
| | - Chang-Song Zhang
- Department of Oromaxillofacial-Head and Neck Surgery, Tianjin Stomatological Hospital, School of Medicine, Nankai University, 75 Dagu Road, Heping District, Tianjin 300041, China; Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, 75 Dagu Road, Heping District, Tianjin 300041, China
| | - Zhao-Yuan Xu
- Department of Oral Medical Center, the First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu 241000, China
| | - Shi-Rui Wang
- Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, 75 Dagu Road, Heping District, Tianjin 300041, China; Department of Operative Dentistry and Endodontics, Tianjin Stomatological Hospital, School of Medicine, Nankai University, 75 Dagu Road, Heping District, Tianjin 300041, China
| | - Tong-Mei Zhang
- Department of Oromaxillofacial-Head and Neck Surgery, Tianjin Stomatological Hospital, School of Medicine, Nankai University, 75 Dagu Road, Heping District, Tianjin 300041, China; Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, 75 Dagu Road, Heping District, Tianjin 300041, China
| | - Yan Zhang
- Department of Oromaxillofacial-Head and Neck Surgery, Tianjin Stomatological Hospital, School of Medicine, Nankai University, 75 Dagu Road, Heping District, Tianjin 300041, China; Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, 75 Dagu Road, Heping District, Tianjin 300041, China.
| | - Su-Xia Liang
- Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, 75 Dagu Road, Heping District, Tianjin 300041, China; Department of Operative Dentistry and Endodontics, Tianjin Stomatological Hospital, School of Medicine, Nankai University, 75 Dagu Road, Heping District, Tianjin 300041, China.
| | - Ying-Bin Yan
- Department of Oromaxillofacial-Head and Neck Surgery, Tianjin Stomatological Hospital, School of Medicine, Nankai University, 75 Dagu Road, Heping District, Tianjin 300041, China; Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, 75 Dagu Road, Heping District, Tianjin 300041, China.
| |
Collapse
|
14
|
Zhou D, Huang Z, Liu J, Tan J, Li H, Ai Y. Gastrodia protects HT22 cells from damage caused by oxygen glucose deprivation and reperfusion through inhibiting ferroptosis. Sci Rep 2025; 15:18470. [PMID: 40425648 PMCID: PMC12117028 DOI: 10.1038/s41598-025-03404-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Accepted: 05/20/2025] [Indexed: 05/29/2025] Open
Abstract
Gastrodin (Gas) is a key active ingredients of Gastrodia elata Bl., with applications in treating cardiovascular and neurodegenerative conditions. However, the impact of Gas on neuronal damage caused by cerebral ischemia/reperfusion remains uncertain. A cell model of oxygen-glucose deprivation/reoxygenation (OGD/R) was established and the viability and apoptosis of HT22 cells were measured using the CCK-8 assay and TUNEL staining. Different kits detected the levels of LDH, Fe2+ and MDA. The levels of ferroptosis-related genes and proteins were evaluated utilizing RT-qPCR and Western blotting. Following OGD/R, there was a decrease in HT22 cell viability and an increase in LDH level and apoptosis rate. Gas (25µM) increased cell viability, decreased LDH, Fe2+, MDA and ACSL4 levels, up-regulated SLC7A11 and GPX4 and ameliorated OGD/R-induced apoptosis (P < 0.01). Ferroptosis inducer Erastin (Era, 10µM) successfully induced ferroptosis in HT22 cells, while Gas treatment attenuated the effect of Era. Era further promoted OGD/R-induced damage and ferroptosis in HT22 cells, whereas Gas inhibited the effect of Era. In conclusion, Gas might provide protection against induced HT22 cell injury caused by OGD/R through inhibiting ferroptosis, shows promising potential for clinical treatment of cerebral ischemia/reperfusion.
Collapse
Affiliation(s)
- Dongyue Zhou
- Jiangxi Province Key Laboratory of Traditional Chinese Medicine Pharmacology, Institute of Traditional Chinese Medicine Health Industry, China Academy of Chinese Medical Sciences, Nanchang, 330115, China
- Jiangxi Health Industry Institute of Traditional Chinese Medicine, Nanchang, 330115, China
| | - Zhixuan Huang
- Jiangxi Province Key Laboratory of Traditional Chinese Medicine Pharmacology, Institute of Traditional Chinese Medicine Health Industry, China Academy of Chinese Medical Sciences, Nanchang, 330115, China
- Jiangxi Health Industry Institute of Traditional Chinese Medicine, Nanchang, 330115, China
| | - Jian Liu
- Jiangxi Province Key Laboratory of Traditional Chinese Medicine Pharmacology, Institute of Traditional Chinese Medicine Health Industry, China Academy of Chinese Medical Sciences, Nanchang, 330115, China
- Jiangxi Health Industry Institute of Traditional Chinese Medicine, Nanchang, 330115, China
| | - Jinlong Tan
- Jiangxi Province Key Laboratory of Traditional Chinese Medicine Pharmacology, Institute of Traditional Chinese Medicine Health Industry, China Academy of Chinese Medical Sciences, Nanchang, 330115, China
- Jiangxi Health Industry Institute of Traditional Chinese Medicine, Nanchang, 330115, China
| | - Hui Li
- Jiangxi Province Key Laboratory of Traditional Chinese Medicine Pharmacology, Institute of Traditional Chinese Medicine Health Industry, China Academy of Chinese Medical Sciences, Nanchang, 330115, China
- Jiangxi Health Industry Institute of Traditional Chinese Medicine, Nanchang, 330115, China
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Yangwen Ai
- Jiangxi Province Key Laboratory of Traditional Chinese Medicine Pharmacology, Institute of Traditional Chinese Medicine Health Industry, China Academy of Chinese Medical Sciences, Nanchang, 330115, China.
- Jiangxi Health Industry Institute of Traditional Chinese Medicine, Nanchang, 330115, China.
| |
Collapse
|
15
|
Yuxuan H, Sixu R, Chenglin L, Xiufen Z, Cuilin Z. Targeting mitochondria quality control for myocardial ischemia-reperfusion injury. Mitochondrion 2025:102046. [PMID: 40419068 DOI: 10.1016/j.mito.2025.102046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 04/24/2025] [Accepted: 05/01/2025] [Indexed: 05/28/2025]
Abstract
Cardiovascular disease (CVD) remains the leading global cause of mortality. Acute myocardial infarction (AMI) refers to acute myocardial ischemia resulting from thrombosis secondary to coronary atherosclerosis, which poses a major threat to human health. Clinically, timely revascularization (reperfusion) represents the basis of clinical treatment for AMI. However, secondary myocardial ischemia-reperfusion injury (MIRI) caused by reperfusion often exacerbates damage, representing a major challenge in clinical practice. Mitochondria represent essential organelles for maintaining cardiac function and cellular bioenergetics in MIRI. In recent years, the role of mitochondrial quality control (MQC) in maintaining cell homeostasis and mediating MIRI has been extensively studied. This review provides a concise overview of MQC mechanisms at the molecular, organelle, and cellular levels and their possible complex regulatory network in MIRI. In addition, potential treatment strategies targeting MQC to mitigate MIRI are summarized, highlighting the gap between current preclinical research and clinical transformation. Overall, this review provides theoretical guidance for further research and clinical translational studies.
Collapse
Affiliation(s)
- He Yuxuan
- Department of Cardiovascular Surgery, The Second Hospital of Jilin University, Changchun 130000, China; Norman Bethune Second Clinical Medical College, Jilin University, Changchun 130000, China
| | - Ren Sixu
- Department of Cardiovascular Surgery, The Second Hospital of Jilin University, Changchun 130000, China; Norman Bethune Second Clinical Medical College, Jilin University, Changchun 130000, China
| | - Liu Chenglin
- China-Japan Union Hospital of Jilin University, Changchun City 130033 Jilin Province, China
| | - Zheng Xiufen
- Department of Surgery, Western University, Ontario, Canada
| | - Zhu Cuilin
- Department of Cardiovascular Surgery, The Second Hospital of Jilin University, Changchun 130000, China; Norman Bethune Second Clinical Medical College, Jilin University, Changchun 130000, China.
| |
Collapse
|
16
|
Huang W, Tang P, Mei J, Zhang Z, Lu G. Identification of molecular subtypes and a prognostic signature based on machine learning and purine metabolism-related genes in breast cancer. Medicine (Baltimore) 2025; 104:e42288. [PMID: 40419914 DOI: 10.1097/md.0000000000042288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/28/2025] Open
Abstract
Breast cancer (BC), one of the most prevalent malignant tumors worldwide, lacks efficacious diagnostic biomarkers and therapeutic targets. This study harnesses multi-omics data to identify novel purine metabolism-related genes (PMRG) as potential biomarkers and risk signatures. Univariate Cox regression was employed to assess the correlation between PMRGs and BC patient prognosis, while a Lasso Cox model was constructed to derive a prognostic signature. Gene set enrichment analysis was utilized to investigate functional differences. Kaplan-Meier survival curves were plotted to examine overall survival disparities between these 2 risk groups, with further exploration into the relationship between the prognostic signature, immune landscape, and drug sensitivity. Ultimately, a predictive nomogram was developed based on these findings. BC patients were stratified into 2 distinct molecular subtypes with significantly different prognoses using the identified PMRG-based signature, which comprised 17 PMRGs. This signature emerged as an independent prognosticator for BC and was integrated into a nomogram along with age, chemotherapy/radiotherapy treatment history, and clinical staging to accurately predict patient outcomes. Moreover, the signature showed associations with the tumor immune microenvironment and drug responsiveness, where lower-risk patients exhibited increased chemotherapeutic sensitivity, immune scores, and decreased tumor purity. Gene set enrichment analysis highlighted significant activation in pathways such as the complement and coagulation cascades, ribosome biogenesis, MAPK signaling, cAMP signaling, and drug metabolism pathways in the low-risk group. The derived PMRG-based signature holds promise for predicting the prognosis of BC patients and guiding their clinical management, including immunotherapy interventions.
Collapse
Affiliation(s)
- Wei Huang
- Department of Surgery, Ninghai Maternal and Child Health Hospital, Ningbo, Zhejiang, China
| | | | | | | | | |
Collapse
|
17
|
Hu XN, Li CF, Huang SM, Nie CL, Pang R. Pescadillo ribosomal biogenesis factor 1 and programmed death-ligand 1 in gastric and head and neck squamous cell carcinoma. World J Gastroenterol 2025; 31:106644. [DOI: 10.3748/wjg.v31.i19.106644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Revised: 04/07/2025] [Accepted: 04/23/2025] [Indexed: 05/21/2025] Open
Abstract
BACKGROUND Gastric cancer (GC) and head and neck squamous cell carcinoma (HNSCC) are common malignancies with high morbidity and mortality rates. Traditional treatments often yield limited efficacy, especially in advanced cases. Recent advancements in immunotherapy, particularly immune checkpoint inhibitors targeting programmed death-ligand 1 (PD-L1), have shown promise. However, the expression and interaction of pescadillo ribosomal biogenesis factor 1 (PES1) and PD-L1 in these cancers remain unclear. Understanding their roles could provide new insights into tumor biology and improve therapeutic strategies.
AIM To investigate the expression levels of PES1 and PD-L1 in tumor tissues of patients with GC and HNSCC.
METHODS A total of 58 cases of GC and HNSCC undergoing surgical resection were selected from January 2022 to January 2024. Paraffin specimens of GC and HNSCC tissues were taken from the patients, and the sections were subjected to staining with immunohistochemistry and hematoxylin-eosin staining, and the protein expression of PES1 and PD-L1 was observed microscopically.
RESULTS Among 58 GC and HNSCC tissues, 30 cases were positive and 28 cases were negative for PES1 expression, and 34 cases were positive and 24 cases were negative for PD-L1 expression. The positive expression rates of PES1 and PD-L1 were 51.72% and 58.62%, respectively. PES1 expression was correlated with the TNM stage, lymph node metastasis, and the depth of infiltration (P < 0.05), and PD-L1 expression was correlated with the differentiation degree, lymph node metastasis, and infiltration depth (P < 0.05).
CONCLUSION PES1 and PD-L1 were positively expressed in GC and HNSCC tissues and correlated with clinical features. They may serve as potential biomarkers for immune-targeted therapies.
Collapse
Affiliation(s)
- Xiao-Nan Hu
- Department of Head and Neck Radiotherapy Ward, Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang Province, China
| | - Chun-Feng Li
- Department of Gastrointestinal Surgical Ward, Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang Province, China
| | - Si-Meng Huang
- Department of Head and Neck Surgery, Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang Province, China
| | - Chun-Lei Nie
- Department of Head and Neck Surgery, Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang Province, China
| | - Rui Pang
- Department of Head and Neck Surgery, Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang Province, China
| |
Collapse
|
18
|
Gao S, Zhou JR, Yokomizo K, Fang J. Nano-drug delivery system of natural products for disease prevention and treatment. Expert Opin Drug Deliv 2025:1-11. [PMID: 40366774 DOI: 10.1080/17425247.2025.2506830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 04/22/2025] [Accepted: 05/12/2025] [Indexed: 05/16/2025]
Abstract
INTRODUCTION Natural products, derived from plants, animals, and microorganisms, offer a wide range of pharmacological activities, including anti-infective, antifungal, anti-tumor, cholesterol-lowering, and anti-inflammatory effects. However, their clinical use is often limited by challenges such as low stability, poor bioavailability, and short half-lives. Thus, developing effective drug delivery systems for these compounds is crucial. AREAS COVERED This review highlights the integration of natural products with nano-drug delivery systems, focusing on recent advancements that utilize the enhanced permeability and retention (EPR) effect to improve their stability, bioavailability, and targeting. By embedding natural compounds into polymeric nanoparticles or similar nanoplatforms, these formulations significantly enhance pharmacokinetic and pharmacodynamic properties, overcoming traditional limitations. EXPERT OPINION Combining natural products with nanoparticle technology shows great potential to expand their therapeutic applications. Although these innovations improve the pharmacological profiles of natural compounds, continued research is essential to optimize clinical use. Advances in nanoparticle design and delivery strategies will be key to maximizing the therapeutic potential of natural products, addressing existing challenges, and enhancing their efficacy in disease treatment.
Collapse
Affiliation(s)
- Shanghui Gao
- Faculty of Pharmaceutical Sciences, Sojo University, Kumamoto, Japan
| | - Jian-Rong Zhou
- Faculty of Pharmaceutical Sciences, Sojo University, Kumamoto, Japan
| | - Kazumi Yokomizo
- Faculty of Pharmaceutical Sciences, Sojo University, Kumamoto, Japan
| | - Jun Fang
- Faculty of Pharmaceutical Sciences, Sojo University, Kumamoto, Japan
| |
Collapse
|
19
|
Chang L, Qin C, Wu J, Jiang H, Xu Q, Chen J, Xu X, Zhang X, Guan M, Deng X. The crosstalk between glutathione metabolism and non-coding RNAs in cancer progression and treatment resistance. Redox Biol 2025; 84:103689. [PMID: 40403492 DOI: 10.1016/j.redox.2025.103689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 05/11/2025] [Accepted: 05/18/2025] [Indexed: 05/24/2025] Open
Abstract
Excessive reactive oxygen species (ROS) are closely associated with the initiation and progression of cancers. As the most abundant intracellular antioxidant, glutathione (GSH) plays a critical role in regulating cellular ROS levels, modulating physiological processes, and is intricately linked to tumor progression and drug resistance. However, the underlying mechanisms remain not fully elucidated. Non-coding RNAs (ncRNAs), including long non-coding RNAs (lncRNAs) and microRNAs (miRNAs), are key regulators of GSH levels. Different ncRNAs modulate various pathways involved in GSH metabolism, and these regulatory targets have the potential to serve as therapeutic targets for enhancing cancer treatment. In this review, we summarize the functions of GSH metabolism and highlight the significance of ncRNA-mediated regulation of GSH in cancer progression, drug resistance, and clinical applications.
Collapse
Affiliation(s)
- Lu Chang
- Department of Laboratory Medicine, Huashan Hospital Fudan University, Shanghai, 200040, China
| | - Chao Qin
- Department of Laboratory Medicine, Huashan Hospital Fudan University, Shanghai, 200040, China
| | - Jianbo Wu
- Department of Laboratory Medicine, Huashan Hospital Fudan University, Shanghai, 200040, China
| | - Haoqin Jiang
- Department of Laboratory Medicine, Huashan Hospital Fudan University, Shanghai, 200040, China
| | - Qianqian Xu
- Department of Laboratory Medicine, Huashan Hospital Fudan University, Shanghai, 200040, China
| | - Jian Chen
- Department of Laboratory Medicine, Huashan Hospital Fudan University, Shanghai, 200040, China
| | - Xiao Xu
- Department of Laboratory Medicine, Huashan Hospital Fudan University, Shanghai, 200040, China
| | - Xinju Zhang
- Department of Laboratory Medicine, Huashan Hospital Fudan University, Shanghai, 200040, China
| | - Ming Guan
- Department of Laboratory Medicine, Huashan Hospital Fudan University, Shanghai, 200040, China.
| | - Xuan Deng
- Department of Laboratory Medicine, Huashan Hospital Fudan University, Shanghai, 200040, China.
| |
Collapse
|
20
|
Zhang X, Sun P, Chu T, Feng Y, Zhang X. Comprehensive bioinformatics analysis of MEX3 family genes in hepatocellular carcinoma. Sci Rep 2025; 15:16971. [PMID: 40374855 PMCID: PMC12081770 DOI: 10.1038/s41598-025-02057-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Accepted: 05/12/2025] [Indexed: 05/18/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is a highly lethal malignancy associated with poor prognosis due to late-stage diagnosis and high recurrence rates. The MEX3 family genes has been implicated in various cancers; however, their roles in HCC remain largely unexplored. This study aims to systematically analyze the expression patterns, prognostic significance, and immune-related functions of MEX3A, MEX3B, MEX3C, and MEX3D in HCC using comprehensive bioinformatics approaches. We conducted a multi-level bioinformatics analysis to investigate the expression, prognostic significance, clinicopathological correlations, genetic alterations, immune associations, and functional mechanisms of MEX3 family members in HCC. Transcriptomic data from TCGA and GEO databases, along with experimental validation via qRT-PCR and Western blotting, were used to assess expression profiles. Kaplan-Meier, ROC curve, and Cox regression analyses were employed for prognostic evaluation. Co-expression, enrichment, and immune infiltration analyses further elucidated the functional and immunological relevance of MEX3 family genes. A prognostic model based on co-expressed genes was constructed and validated using LASSO and time-dependent ROC analyses. MEX3A, MEX3B, MEX3C, and MEX3D were significantly upregulated in HCC tissues compared to normal liver tissues (P < 0.05). ROC curve analysis demonstrated high diagnostic accuracy, particularly for MEX3A (AUC = 0.915). Kaplan-Meier survival analysis indicated that elevated MEX3A and MEX3C expression was associated with poorer overall survival (OS) and disease-specific survival (DSS) (P < 0.05). Mutation analysis revealed that MEX3A exhibited the highest alteration frequency (11%), primarily through gene amplifications. Immune infiltration analysis demonstrated significant correlations between MEX3 expression and multiple immune cell populations, including regulatory T cells (Tregs), cytotoxic T cells, and macrophages. Moreover, MEX3B, MEX3C, and MEX3D expression correlated with key immune checkpoint genes, including PDCD1, CD274, and CTLA4. Functional enrichment analysis revealed that MEX3 co-expressed genes were significantly involved in RNA metabolism, immune response regulation, and oncogenic signaling pathways. A 17-gene MEX3 co-expression-based prognostic model stratified patients into high- and low-risk groups with significantly different survival outcomes (AUC = 0.791 at 1 year). This study highlights the oncogenic potential of MEX3 family members in HCC and their associations with immune regulation. The findings suggest that MEX3 family genes could serve as potential biomarkers for HCC prognosis and immunotherapy responsiveness. Further experimental validation is warranted to elucidate the mechanistic roles of MEX3 family genes in HCC progression and immune evasion.
Collapse
Affiliation(s)
- Xuezhong Zhang
- Department of Laboratory Medicine, Zibo Central Hospital, Zibo, Shandong, China
| | - Peng Sun
- Department of Gastroenterology, Zibo Central Hospital, Zibo, Shandong, China
| | - Tingting Chu
- Department of Laboratory Medicine, Zibo Central Hospital, Zibo, Shandong, China
| | - Yuling Feng
- Department of Infection Disease and Hepatology Ward, Zibo Central Hospital, Zibo, Shandong, China.
| | - Xuebin Zhang
- Department of Anorectal Surgery, Dongying People's Hospital (Dongying Hospital of Shandong Provincial Hospital Group), Dongying, China.
| |
Collapse
|
21
|
Rafaqat S, Noshair I, Shahid M, Bibi S, Hafeez R, Hamid H. Correlation between prognostic markers and clinical parameters in hepatocellular carcinoma: Pathophysiological aspects to therapeutic targets. World J Gastrointest Oncol 2025; 17:106278. [DOI: 10.4251/wjgo.v17.i5.106278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 03/08/2025] [Accepted: 03/17/2025] [Indexed: 05/15/2025] Open
Abstract
One of the main causes of cancer-related morbidity and mortality globally is hepatocellular carcinoma (HCC). At every stage of the disease, HCC may now be treated using a variety of therapy techniques. Nevertheless, despite the abundance of effective therapeutic choices, the prognosis for patients with HCC is still typically dismal. Prognostic indicators are crucial when assessing prognosis and tracking tumor metastases or recurrence. There are many prognostic markers in HCC. We mainly focused on newly reported prognostic markers such as MEX3A, apolipoprotein B, alpha-fetoprotein, circulating tumor cells, SAMD13, Agrin, and Glypican-3 in the pathogenesis of HCC. Further, we highlighted how these prognostic markers correlated to clinical parameters such as tumor node metastasis, tumor diameter, differentiation, hepatocirrhosis, vascular invasion, and others in HCC. Therefore, identifying specific prognostic biomarkers of HCC helps to provide a great opportunity to improve the prognosis in patients with HCC and provide therapeutic targets.
Collapse
Affiliation(s)
- Saira Rafaqat
- Department of Zoology, Lahore College for Women University, Lahore 54000, Pakistan
| | - Iqra Noshair
- Department of Zoology, Lahore College for Women University, Lahore 54000, Pakistan
| | - Momina Shahid
- Department of Zoology, University of Narowal, Narowal 54000, Pakistan
| | - Sadaf Bibi
- Department of Zoology, Government College University, Lahore 54000, Pakistan
| | - Ramsha Hafeez
- Department of Zoology, Lahore College for Women University, Lahore 54000, Pakistan
| | - Hafsa Hamid
- Department of Biotechnology, Lahore College for Women University, Lahore 54000, Pakistan
| |
Collapse
|
22
|
Saimaiti W, Ma J, Dilimulati P, Wang Y. Sunitinib-resistant renal cell carcinoma cell-derived exosomes promote facilitation of tumor progression via secretion of the lncRNA SNHG16. Hum Cell 2025; 38:100. [PMID: 40358820 DOI: 10.1007/s13577-025-01228-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Accepted: 05/01/2025] [Indexed: 05/15/2025]
Abstract
Renal cell carcinoma (RCC) is one of the most common tumors of high malignancy in the urological system. Sunitinib is commonly used to treat RCC, while drug resistance severely limited the therapeutic efficacy. Tumor-derived exosomes play important roles in facilitating cancer development. However, the role of drug-resistant tumor-derived exosomes in tumorigenesis and resistance of RCC has not been elucidated. Here we isolated sunitinib-sensitive/resistant RCC cells-derived exosomes, characterized by transmission electron microscopy (TEM) and western blot. Furthermore, co-culture experiments were performed and we found that sunitinib-resistant RCC cells-derived exosomes (R-exos) promoted cell proliferation and upregulated proliferation-related genes cyclin D1 (CCND1) and proliferating cell nuclear antigen (PCNA) expression, and inhibited apoptosis and the expression of Bax and Caspase-3 of sunitinib-resistant RCC (RCC/R) cells by delivering lncRNA small nuclear RNA host gene 16 (SNHG16). In resistant cell-derived xenograft (CDX-R) models, R-exos induced tumor growth in vivo, while knockdown of SNHG16 effectively diminished the tumorigenesis of RCC. Moreover, SNHG16 positively regulated the expression of trophinin associated protein (TROAP) by sponging miR-106a-5p in RCC cells, whereas inhibition of miR-106a-5p or overexpression of TROAP greatly reversed the suppression of tumorigenesis and sunitinib resistant by silencing SNHG16. R-exos lncRNA SNHG16 promoted sunitinib resistant and malignant progress by regulating the miR-106a-5p/TROAP axis, and targeting SNHG16/miR-106a-5p/TROAP axis may be a novel therapeutic approach for sunitinib-treated patients of RCC.
Collapse
Affiliation(s)
- WeiLijiang Saimaiti
- Department of Pediatric Urology, First Affiliated Hospital, Xinjiang Medical University, Urumqi, 830054, China
| | - Jun Ma
- Department of Urology, First Affiliated Hospital, Xinjiang Medical University, No. 137, Liyuushan South Road, Urumqi, 830054, Xinjiang, China
| | - Paluoke Dilimulati
- Department of Pediatric Urology, First Affiliated Hospital, Xinjiang Medical University, Urumqi, 830054, China
| | - Yujie Wang
- Department of Urology, First Affiliated Hospital, Xinjiang Medical University, No. 137, Liyuushan South Road, Urumqi, 830054, Xinjiang, China.
| |
Collapse
|
23
|
Zhu J, Yu W. Association Between Bariatric Surgery and Kidney Stone Risk: Addressing Limitations and Future Research Directions. Obes Surg 2025:10.1007/s11695-025-07913-6. [PMID: 40341488 DOI: 10.1007/s11695-025-07913-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2025] [Revised: 05/01/2025] [Accepted: 05/05/2025] [Indexed: 05/10/2025]
Affiliation(s)
- Junrou Zhu
- Zhejiang Chinese Medical University, Hangzhou, China
- Hangzhou First People's Hospital, Hangzhou, China
| | - Wenhua Yu
- Hangzhou First People's Hospital, Hangzhou, China.
| |
Collapse
|
24
|
Liu S, Jiang Q, Li W, Shi J, Wu B, Xiong M, Huang L. Identification and Validation of Tryptophan Metabolism-Related Genes in Diabetic Kidney Disease and Construction of a Clinical Prediction Model. J Diabetes Res 2025; 2025:2736801. [PMID: 40376698 PMCID: PMC12081153 DOI: 10.1155/jdr/2736801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Accepted: 04/08/2025] [Indexed: 05/18/2025] Open
Abstract
Background: Diabetic kidney disease (DKD) is a common microvascular complication of diabetes mellitus (DM). Amino acid (AA) homeostasis has an important impact on renal hemodynamics and glomerular hyperfiltration in patients with DKD, and the metabolite level of tryptophan (TRP), an AA, has been associated with various diseases. Methods: In this study, DKD tubule- and glomerulus-related microarray datasets were collected from the GEO database, and DKD-related modular genes were identified by weighted gene coexpression network analysis (WGCNA). TRP metabolism-related genes (TRGs) were downloaded from the MSigDB database, and the key genes were obtained by taking the intersection of DKD differentially expressed genes, TRGs, and modular genes. Validated with the Nephrseq v5 database and performed clinical prediction model construction. The association of pivotal genes with immune cell infiltration was verified using CIBERSORTx software. The protein expression of the key genes was verified by qPCR, Western blot, immunohistochemistry, and immunofluorescence. Results: Four hundred and seventy seven DEGs were identified in the GSE30529 dataset, 392 DEGs were identified in the GSE30528 dataset, and the intersection of the DEGs in the two datasets, the module with the most significant correlation with DKD obtained by WGCNA, and the TRGs were taken, respectively. Five key genes were finally obtained (AOC1, HAAO, STAT1, OGDHL, and TDO2). Compared with control-group mice, the expression of AOC1, HAAO, and OGDHL was significantly downregulated, and the expression of STAT1 and TDO2 was significantly elevated in DKD mice. The diagnostic model was constructed using the key genes AUC = 0.996. Conclusion: Our study suggests that the AOC1, HAAO, and STAT1 genes may be potential diagnostic biomarkers of tubular injury in DKD. OGDHL and TDO2 may be potential diagnostic biomarkers of glomerular injury in DKD. The model constructed using AOC1, HAAO, STAT1, OGDHL, and TDO2 had good disease differentiation.
Collapse
Affiliation(s)
- Shaojie Liu
- Department of Nephrology, Blood Purification Research Centre, Ningde Hospital of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Ningde, China
| | - Qingqing Jiang
- Department of Nephrology, Blood Purification Research Centre, Ningde Hospital of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Ningde, China
| | - Wenli Li
- Department of Nephrology, Blood Purification Research Centre, Ningde Hospital of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Ningde, China
| | - Jinbao Shi
- Department of Nephrology, Blood Purification Research Centre, Ningde Hospital of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Ningde, China
| | - Binxuan Wu
- Department of Nephrology, Blood Purification Research Centre, Ningde Hospital of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Ningde, China
| | - Man Xiong
- Department of Nephrology, Blood Purification Research Centre, Ningde Hospital of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Ningde, China
| | - Liuying Huang
- Department of Nephrology, Blood Purification Research Centre, Ningde Hospital of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Ningde, China
| |
Collapse
|
25
|
Wilczak M, Surman M, Jankowska U, Skupien-Rabian B, Przybyło M. MGAT3 and MGAT5 overexpression alters the protein cargo of extracellular vesicles released by metastatic melanoma cells. Biochem Biophys Res Commun 2025; 762:151749. [PMID: 40199132 DOI: 10.1016/j.bbrc.2025.151749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 03/25/2025] [Accepted: 04/01/2025] [Indexed: 04/10/2025]
Abstract
Extracellular vesicles (EVs) are potential non-invasive diagnostic, prognostic and therapeutic tools. Additionally, they are important contributors to tumorigenesis. Glycosylation has been found to modulate the composition of the EV proteome. Increased amounts of β1,6-branched N-glycans, synthesized by N-acetylglucosaminyltransferase V (GnT-V), are most commonly observed in melanoma and are associated with decreased cell adhesion and increased metastasis. The opposite effect is caused by the addition of bisecting GlcNAc by N-acetylglucosaminyltransferase III (GnT-III). To date, the impact of these enzymes on EV cargo in melanoma remains unexplored. Flow cytometry was used to study the surface glycosylation of genetic variants of WM266-4 melanoma cells with induced overexpression of GnT-III or GnT-V encoding genes (MGAT3 or MGAT5) and EVs released by these cells. LC-MS/MS proteomics was applied to analyze the effect of altered glycosylation on the proteome of released EVs, followed by detailed bioinformatic analysis. Flow cytometry analysis revealed dynamic changes in the surface glycosylation of EVs derived from melanoma cells overexpressing MGAT3 or MGAT5. Induced overexpression of MGAT3 or MGAT5 also caused significant changes in the proteome of EVs. The proteomic analysis identified a total of 1770 microvesicular and 704 exosomal proteins that play different roles in melanoma progression, including those with established diagnostic/prognostic potential and those closely associated with melanoma onset. Proteomic profiling of EVs derived from cells overexpressing MGAT3 and MGAT5 revealed functional changes in EV protein content driven by glycosylation modifications. The study presented a potential multifaced application of melanoma-derived EVs for diagnostic and prognostic purposes.
Collapse
Affiliation(s)
- Magdalena Wilczak
- Department of Glycoconjugate Biochemistry, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, 30-387, Krakow, Poland; Doctoral School of Exact and Natural Sciences, Jagiellonian University, 30-348, Krakow, Poland.
| | - Magdalena Surman
- Department of Glycoconjugate Biochemistry, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, 30-387, Krakow, Poland.
| | - Urszula Jankowska
- Proteomics and Mass Spectrometry Core Facility, Malopolska Centre of Biotechnology, Jagiellonian University, 30-387, Krakow, Poland.
| | - Bozena Skupien-Rabian
- Proteomics and Mass Spectrometry Core Facility, Malopolska Centre of Biotechnology, Jagiellonian University, 30-387, Krakow, Poland.
| | - Małgorzata Przybyło
- Department of Glycoconjugate Biochemistry, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, 30-387, Krakow, Poland.
| |
Collapse
|
26
|
Kuang Y, Zhao Y, Miao Z, Xu Y, Yang Q. Oxaloacetate stimulates phosphorylation of epidermal growth factor receptor in epithelial cells in vitro. Growth Factors 2025:1-13. [PMID: 40314117 DOI: 10.1080/08977194.2025.2499634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 04/16/2025] [Indexed: 05/03/2025]
Abstract
Oxaloacetate (OA) is a pivotal endogenous metabolite. Within our investigation, we ascertained that OA functions as an agonist for the epidermal growth factor receptor (EGFR), a key protagonist in the genesis of diverse tumours. We substantiated that escalating concentrations of OA initially enhanced the cellular viability of several cancer cells, followed by subsequent attenuation, which is similar to the effect of EGF. Furthermore, the protein phosphorylation profile in HepG2 cells exposed to OA closely paralleled that induced by epidermal growth factor (EGF). Additional findings underscored the capability of OA to induce the generation of EGFR dimers. Finally, our observations revealed that OA governs the activation of AKT and Erk, the typical downstream signalling proteins of EGFR. We postulate that the endogenous metabolite OA can function as either an agonist or inhibitor of EGFR at specific concentrations to modulate tumour proliferation, and provide new insights into the regulation of EGFR activation.
Collapse
Affiliation(s)
- Ye Kuang
- Department of Pathogenobiology, College of Basic Medical Sciences, Jilin University, Changchun City, China
| | - Yuxiang Zhao
- Department of Pathogenobiology, College of Basic Medical Sciences, Jilin University, Changchun City, China
| | - Zeyu Miao
- Department of Pathogenobiology, College of Basic Medical Sciences, Jilin University, Changchun City, China
| | - Yang Xu
- Department of Pathogenobiology, College of Basic Medical Sciences, Jilin University, Changchun City, China
| | - Qing Yang
- Department of Pathogenobiology, College of Basic Medical Sciences, Jilin University, Changchun City, China
| |
Collapse
|
27
|
Leng J, Sun J, Guo Z, Wang L, Song L. The molecular mechanism of ferroptosis in the Pacific oyster Crassostrea gigas under Erastin treatment or high temperature stress. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2025; 166:105366. [PMID: 40185349 DOI: 10.1016/j.dci.2025.105366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 02/25/2025] [Accepted: 03/29/2025] [Indexed: 04/07/2025]
Abstract
Ferroptosis is an iron- and lipotoxicity-dependent form of programmed cell death, and it is distinct from apoptosis, pyroptosis, and autophagy. In the present study, the hemocytes were found to be shrunken under Erastin treatment or high temperature stress. The mitochondrial atrophy, crest loss and fracture were observed in hemocytes under high temperature stress. In addition, the fluorescence intensity of mitochondrial probe JC-1 monomers increased significantly in hemocytes under high temperature stress. Hemocytes were found to be wrinkled under ultrastructure and the contents of LPO, ROS and GSH increased significantly under Erastin treatment or high temperature stress. The band intensity of CgVDAC2 also decreased under Erastin treatment or high temperature stress. The mRNA expressions of genes involved in enhancing the antioxidation system as well as genes involved in promoting the iron metabolism all decreased significantly under Erastin treatment or high temperature stress. Those of genes involved in impairing the antioxidation system, genes involved in inhibiting the iron metabolism, as well as genes involved in reducing the lipid peroxidation all increased significantly under Erastin treatment or high temperature stress. These results indicated that Erastin could activate the three key ferroptotic signaling pathways in oyster and the activation mechanism of ferroptosis in oyster under high temperature stress was similar with that under Erastin treatment.
Collapse
Affiliation(s)
- Jinyuan Leng
- College of Life Sciences, Liaoning Normal University, Dalian, 116029, Liaoning, China; Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Liaoning Key Laboratory of Marine Animal Immunology & Disease Control, Dalian Ocean University, Dalian, 116023, China
| | - Jiejie Sun
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Liaoning Key Laboratory of Marine Animal Immunology & Disease Control, Dalian Ocean University, Dalian, 116023, China.
| | - Zhicheng Guo
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Liaoning Key Laboratory of Marine Animal Immunology & Disease Control, Dalian Ocean University, Dalian, 116023, China
| | - Lingling Wang
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Laboratory of Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266235, China; Liaoning Key Laboratory of Marine Animal Immunology & Disease Control, Dalian Ocean University, Dalian, 116023, China
| | - Linsheng Song
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Laboratory of Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266235, China; Liaoning Key Laboratory of Marine Animal Immunology & Disease Control, Dalian Ocean University, Dalian, 116023, China.
| |
Collapse
|
28
|
Geng Y, Luo K, Stam J, Oosterhuis D, Gorter AR, van den Heuvel M, Crescitelli R, de Meijer VE, Wolters JC, Olinga P. Characterization of Extracellular Vesicles Derived From Human Precision-Cut Liver Slices in Metabolic Dysfunction-Associated Steatotic Liver Disease. JOURNAL OF EXTRACELLULAR BIOLOGY 2025; 4:e70043. [PMID: 40313415 PMCID: PMC12042696 DOI: 10.1002/jex2.70043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 01/30/2025] [Accepted: 02/28/2025] [Indexed: 05/03/2025]
Abstract
Extracellular vesicles (EVs) are cell-produced, membrane-surrounded vesicles that harbour the biological features of donor cells. In the current study, we are the first to isolate and characterize EVs isolated from human precision-cut liver slices (PCLS), obtained from both healthy and metabolic dysfunction-associated steatohepatitis (MASH) cirrhotic livers. PCLS derived from patients can faithfully represent disease conditions in humans. EVs were isolated from human PCLS after incubating in normal medium or modified medium that mimics the pathophysiological environment of metabolic dysfunction associated liver disease (MASLD). MASH PCLS produced higher amounts of EVs compared to healthy PCLS (p < 0.001). Mass spectrometry revealed that around 300 proteins were significantly different in EVs derived from MASH PCLS versus healthy PCLS (FDR < 0.05), irrespective of the type of medium. Significantly changed EV proteins were largely involved in signalling receptor binding function and showed potential in promoting fibrosis. In the liver, these ligand-associated receptors are highly expressed in hepatic stellate cells, and the MASH EVs functionally promoted the activation of hepatic stellate cells. Furthermore, the amounts of EpCAM and ITGA3 in EVs were positively associated with the progression of MASLD, which suggests the use of liver-derived EVs as potential biomarkers for MASLD. Characterization of EVs derived from human PCLS may assist future studies in investigating the pathogenesis and identifying liver-specific EVs as biomarkers of MASLD.
Collapse
Affiliation(s)
- Yana Geng
- Department of Pharmaceutical Technology and Biopharmacy, Groningen Research Institute of PharmacyUniversity of GroningenGroningenthe Netherlands
| | - Ke Luo
- Department of Pharmaceutical Technology and Biopharmacy, Groningen Research Institute of PharmacyUniversity of GroningenGroningenthe Netherlands
| | - Janine Stam
- Department of Pharmaceutical Technology and Biopharmacy, Groningen Research Institute of PharmacyUniversity of GroningenGroningenthe Netherlands
- Department of Analytical Biochemistry, Groningen Research Institute of PharmacyUniversity of GroningenGroningenthe Netherlands
| | - Dorenda Oosterhuis
- Department of Pharmaceutical Technology and Biopharmacy, Groningen Research Institute of PharmacyUniversity of GroningenGroningenthe Netherlands
| | - Alan R. Gorter
- Department of Pharmaceutical Technology and Biopharmacy, Groningen Research Institute of PharmacyUniversity of GroningenGroningenthe Netherlands
| | - Marius van den Heuvel
- Division of Pathology, Department of Pathology and Medical BiologyUniversity of Groningen, University Medical Center GroningenGroningenthe Netherlands
| | - Rossella Crescitelli
- Department of Surgery, Sahlgrenska Center for Cancer Research and Wallenberg Centre for Molecular and Translational Medicine, Institute of Clinical SciencesSahlgrenska Academy, University of GothenburgGöteborgSweden
| | - Vincent E. de Meijer
- Department of Surgery, Section of Hepatobiliary Surgery & Liver TransplantationUniversity of Groningen, University Medical Center GroningenGroningenthe Netherlands
| | - Justina C. Wolters
- Department of PediatricsUniversity Medical Center Groningen, University of GroningenGroningenthe Netherlands
| | - Peter Olinga
- Department of Pharmaceutical Technology and Biopharmacy, Groningen Research Institute of PharmacyUniversity of GroningenGroningenthe Netherlands
| |
Collapse
|
29
|
Lei Y, Dong R, Sun C, Hu Y, Yan Y, Song G, Wang Y. The Role of CXCL13 in GC-1 Cell Cycle Arrest Induced by Titanium Dioxide Nanoparticles Through JAK2/STAT3 Signaling Pathway. J Appl Toxicol 2025; 45:830-840. [PMID: 39777719 DOI: 10.1002/jat.4747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 12/02/2024] [Accepted: 12/07/2024] [Indexed: 01/11/2025]
Abstract
Titanium dioxide nanoparticles (TiO2 NPs) can induce the cell cycle arrest in spermatogonia, and the JAK2/STAT3 signaling pathway plays a pivotal role in cell cycle progression, but the specific upstream regulatory mechanisms are not completely clarified. The purpose of this study was to investigate whether CXCL13 regulated the JAK2/STAT3 signaling pathway to participate in cell cycle arrest after mouse spermatogonia cell line (GC-1) exposure to TiO2 NPs. The GC-1 cells were treated with TiO2 NPs at different concentrations (0, 10, 20, 30, and 40 μg/mL) for 24 h to detect cell viability, cell cycle distribution, CXCL13 protein, JAK2/STAT3 pathway-related proteins, and cell cycle-related proteins. The CXCL13 recombinant protein was used to verify the role of CXCL13 in cell cycle and JAK2/STAT3 signaling pathway. TiO2 NPs inhibited cell viability; regulated cell cycle-related proteins including remarkably decreased Cyclin D1, CDK4, Cyclin E1, and CDK2 as well as increased p21; and induced cell cycle arrest at the G0/G1 phase. TiO2 NPs inhibited the levels of CXCL13 protein and weakened the activation of the JAK2/STAT3 signaling pathway by reducing the levels of p-JAK2/JAK2 and p-STAT3/STAT3 proteins. Furthermore, CXCL13 mitigated the suppression of the JAK2/STAT3 signaling pathway and the G0/G1 cell cycle arrest caused by TiO2 NPs. Taken together, TiO2 NPs downregulated the expression of CXCL13 to inhibit the activation of downstream JAK2/STAT3 signaling pathway, eventually inducing cell cycle arrest at the G0/G1 phase. These results provide a novel insight for complemented understanding of the mechanisms of TiO2 NPs-induced cell cycle arrest in GC-1 cells.
Collapse
Affiliation(s)
- Yuzhu Lei
- Department of Preventive Medicine/the Key Laboratory for Prevention and Control of Emerging Infectious Diseases and Public Health Security, the Xinjiang Production and Construction Corps, School of Medicine, Shihezi University, Shihezi, Xinjiang, China
| | - Ruoyun Dong
- Xi'an Center for Disease Control and Prevention, Xi'an, China
| | - Chenhao Sun
- Department of Preventive Medicine/the Key Laboratory for Prevention and Control of Emerging Infectious Diseases and Public Health Security, the Xinjiang Production and Construction Corps, School of Medicine, Shihezi University, Shihezi, Xinjiang, China
| | - Yunhua Hu
- Department of Preventive Medicine/the Key Laboratory for Prevention and Control of Emerging Infectious Diseases and Public Health Security, the Xinjiang Production and Construction Corps, School of Medicine, Shihezi University, Shihezi, Xinjiang, China
| | - Yizhong Yan
- Department of Preventive Medicine/the Key Laboratory for Prevention and Control of Emerging Infectious Diseases and Public Health Security, the Xinjiang Production and Construction Corps, School of Medicine, Shihezi University, Shihezi, Xinjiang, China
| | - Guanling Song
- Department of Preventive Medicine/the Key Laboratory for Prevention and Control of Emerging Infectious Diseases and Public Health Security, the Xinjiang Production and Construction Corps, School of Medicine, Shihezi University, Shihezi, Xinjiang, China
| | - Yan Wang
- School of Medicine, Shihezi University, Shihezi, Xinjiang, China
| |
Collapse
|
30
|
Pourhabib Mamaghani M, Mousavikia SN, Azimian H. Ferroptosis in cancer: Mechanisms, therapeutic strategies, and clinical implications. Pathol Res Pract 2025; 269:155907. [PMID: 40101548 DOI: 10.1016/j.prp.2025.155907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 02/10/2025] [Accepted: 03/08/2025] [Indexed: 03/20/2025]
Abstract
The resistance of cancer cells to existing treatments has become a major challenge for researchers despite advancements in cancer treatment. Studies have shown that this resistance is due to cancer cells evading apoptosis. Moreover, the most common form of cell death induced by chemotherapy and radiotherapy is apoptosis. One of the most essential mechanisms cancer cells escape apoptosis is the excessive expression of tumors' apoptosis inhibitors. Therefore, finding a non-apoptotic pathway that bypasses apoptosis could be a hopeful strategy for cancer treatment. Ferroptosis has been identified as a non-apoptotic and regulated cell death process characterized by the accumulation of lipid peroxides and iron-dependent reactive oxygen species (ROS). Although studies have shown that ferroptosis plays a role in the development of many diseases, including cancer, it also has the potential to decrease resistance to current treatments, such as chemotherapy. Additionally, research has shown that ferroptosis successfully kills cancer cells, such as breast, stem, and lung cancer cells. Therefore, ferroptosis can be identified as a beneficial therapeutic mechanism for cancer treatment. Although ferroptosis has been introduced as an effective treatment path for cancer, its role, along with its therapeutic inducers, in increasing the therapeutic effect has not been investigated. In this review, we aim to introduce ferroptosis, compare it with other cell deaths known so far, and explain its role in cancer treatment. We believe that ferroptosis can be widely used to overcome cancer cells.
Collapse
Affiliation(s)
- Mina Pourhabib Mamaghani
- Department of Medical Physics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyedeh Nasibeh Mousavikia
- Department of Medical Physics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Medical Physics Research Center, Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hosein Azimian
- Department of Medical Physics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Medical Physics Research Center, Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
31
|
Han W, Bian X, Fu H, Liu M, Wang H, Liu H. Integrating Single-Cell Sequencing and Transcriptome Analysis to Investigate the Role of Ferroptosis in Ischemic Stroke and the Molecular Mechanisms of Immune Checkpoints. World Neurosurg 2025; 197:123908. [PMID: 40118372 DOI: 10.1016/j.wneu.2025.123908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Revised: 03/10/2025] [Accepted: 03/11/2025] [Indexed: 03/23/2025]
Abstract
BACKGROUND Early diagnosis of ischemic stroke (IS) remains challenging. Given the crucial role of ferroptosis in IS, this study aims to identify key genes associated with ferroptosis in IS, providing insights into its molecular mechanisms and potential biomarkers for early detection. METHODS The single-cell transcriptome dataset GSE247474 from the Gene Expression Omnibus. Ferroptosis scores in astrocytes were calculated using the WP_FERROPTOSIS gene set, and differential analysis was conducted to compare ferroptosis activity between the disease and control groups. Key ferroptosis-related genes were identified using Lasso regression and support vector machine algorithms, and their diagnostic potential was assessed through receiver operating characteristic curve analysis. Additionally, we performed immune infiltration analysis and transcription factor network prediction. Pseudotime analysis was used to explore the differentiation trajectories of astrocytes and T-cell subsets. RESULTS Astrocytes in the disease group showed significantly higher ferroptosis scores than those in the control group. Using machine learning algorithms, we identified 3 key ferroptosis-related genes-SLC3A2 (solute carrier family 3 member 2), FDFT1 (farnesyl-diphosphate farnesyltransferase 1), and BACH1 (BTB and CNC homology 1)-and validated their diagnostic value (area under the curve >0.9). Immune infiltration analysis revealed that SLC3A2 and BACH1 expression levels were positively correlated with CD4+ follicular T cells and negatively correlated with CD4+ memory T cells. FDFT1 showed positive correlations with both mast cells and CD4+ memory T cells. Pseudotime analysis demonstrated dynamic changes in key gene expression along the differentiation trajectories of astrocytes and T cells. CONCLUSIONS SLC3A2, FDFT1, and BACH1 are potential molecular markers for IS diagnosis.
Collapse
Affiliation(s)
- Weidong Han
- Department of Clinical Laboratory, Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), Nantong, Jiangsu, China
| | - Xiaonan Bian
- Department of Clinical Laboratory, Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), Nantong, Jiangsu, China
| | - Haiyang Fu
- Department of Neurobiology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Min Liu
- Department of Clinical Laboratory, Liupanshui Maternal and Child Health Hospital, Liupanshui, Guizhou, China
| | - Hongliang Wang
- Department of Clinical Laboratory, Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), Nantong, Jiangsu, China
| | - Haimei Liu
- Department of Clinical Laboratory, Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), Nantong, Jiangsu, China.
| |
Collapse
|
32
|
Lin C, Xie S, Wang M, Shen J. PRDX1 knockdown promotes erastin-induced ferroptosis and impedes diffuse large B-cell lymphoma development by inhibiting the MAPK/ERK pathway. BMC Cancer 2025; 25:806. [PMID: 40307771 PMCID: PMC12042503 DOI: 10.1186/s12885-025-14173-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 04/16/2025] [Indexed: 05/02/2025] Open
Abstract
AIM Diffuse large B-cell lymphoma (DLBCL) is an aggressive lymphoma and DLBCL cells are highly sensitive to ferroptosis. The purpose of this research was to evaluate the role and molecular mechanism of peroxiredoxin 1 (PRDX1) on ferroptosis in DLBCL. METHODS The expression of PRDX1 in DLBCL tissues and cells was detected using bioinformatics analysis and reverse transcription quantitative PCR. The impacts of PRDX1 on DLBCL cell proliferation, apoptosis, migration, invasion, and ferroptosis were assessed through a series of in vitro experiments. A xenograft tumor model was constructed to verify the roles of PRDX1 in vivo. Transcriptome sequencing was conducted to identify PRDX1-mediated signaling pathways. Anisomycin, an agonist of mitogen-activated protein kinase (MAPK), was used to explore the modulation of PRDX1 on the MAPK pathway. RESULTS PRDX1 expression was upregulated in DLBCL. PRDX1 knockdown inhibited DLBCL cell proliferation, migration, and invasion, promoted apoptosis, and suppressed xenograft tumor growth. PRDX1 knockdown boosted erastin-induced ferroptosis by increasing the levels of iron and MDA, while decreasing the levels of GSH. It also promoted COX2 protein expression and inhibited GPX4 and SLC7A11 protein levels. PRDX1 knockdown reduced the phosphorylation levels of MEK and ERK both under conditions with or without erastin induction. The MAPK/ERK pathway agonist anisomycin, significantly reversed the inhibitory effects of PRDX1 knockdown on the malignant behaviors of DLBCL cells and the promotion of ferroptosis. CONCLUSION PRDX1 knockdown facilitates erastin-induced ferroptosis and obstacles DLBCL progression by inhibiting the MAPK/ERK pathway, offering a potential treatment strategy for DLBCL treatment.
Collapse
MESH Headings
- Ferroptosis/drug effects
- Ferroptosis/genetics
- Humans
- Lymphoma, Large B-Cell, Diffuse/pathology
- Lymphoma, Large B-Cell, Diffuse/genetics
- Lymphoma, Large B-Cell, Diffuse/metabolism
- Lymphoma, Large B-Cell, Diffuse/drug therapy
- Animals
- Mice
- Peroxiredoxins/genetics
- Peroxiredoxins/metabolism
- MAP Kinase Signaling System/drug effects
- MAP Kinase Signaling System/genetics
- Piperazines/pharmacology
- Cell Proliferation/drug effects
- Cell Line, Tumor
- Xenograft Model Antitumor Assays
- Cell Movement
- Gene Knockdown Techniques
- Gene Expression Regulation, Neoplastic
- Apoptosis/drug effects
- Male
- Female
Collapse
Affiliation(s)
- Chuanming Lin
- Department of Hematology, First Affiliated Hospital of Gannan Medical University, No. 128, Jinling Road, Economic Development District, Ganzhou City, Jiangxi Province, 341000, China
- Fujian Medical University Union Hospital, No. 29, Xinquan Road, Gulou District, Fuzhou City, Fujian Province, 350001, China
| | - Shuiling Xie
- Department of Hematology, First Affiliated Hospital of Gannan Medical University, No. 128, Jinling Road, Economic Development District, Ganzhou City, Jiangxi Province, 341000, China
| | - Menger Wang
- Gannan Medical University, Rongjiang New District University Park, Ganzhou City, Jiangxi Province, 341099, China
| | - Jianzhen Shen
- Fujian Medical University Union Hospital, No. 29, Xinquan Road, Gulou District, Fuzhou City, Fujian Province, 350001, China.
| |
Collapse
|
33
|
Kwun MS, Lee DG. Ferroptosis-Like Death Induction in Saccharomyces cerevisiae by Gold Nanoparticles. J Microbiol Biotechnol 2025; 35:e2501029. [PMID: 40295204 PMCID: PMC12089944 DOI: 10.4014/jmb.2501.01029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Revised: 02/11/2025] [Accepted: 02/24/2025] [Indexed: 04/30/2025]
Abstract
Ferroptosis, a novel form of regulated cell death (RCD), has emerged as a promising therapeutic strategy for cancer treatment. While gold nanoparticles (AuNPs) are known to induce cell death and ferroptosis in combination with certain antibiotics, the mechanisms underlying ferroptosis in microorganisms remain poorly understood. This study aimed to investigate whether AuNPs induce ferroptosis-like cell death in the eukaryotic microbe Saccharomyces cerevisiae. Our findings revealed that AuNPs significantly reduced cell viability in S. cerevisiae, suggesting their ability to trigger cell death. Ferroptosis-related precursors, including intracellular iron overload and depletion of glutathione (GSH), were observed, leading to the inactivation of glutathione peroxidase (GPx). These changes were associated with the accumulation of reactive oxygen species (ROS) and lipid peroxidation, which amplified oxidative stress within the cells. Elevated ROS levels and lipid peroxidation further resulted in membrane rupture and the formation of 8-hydroxydeoxyguanosine, indicating DNA damage. Mitochondrial dysfunction, a hallmark of ferroptosis, was also evident. AuNP treatment caused mitochondrial membrane potential hyperpolarization and a reduction in mitochondrial membrane density. Unlike previously characterized forms of RCD, ferroptosis-like death in S. cerevisiae did not involve chromatin condensation, DNA fragmentation, or metacaspase activation. Finally, ferroptosis-like characteristics were confirmed using Liperfluo, a lipid ROS-specific probe. In conclusion, this study demonstrated that AuNPs can induce ferroptosis-like cell death in S. cerevisiae. These findings highlight the potential of AuNPs as antifungal agents and contribute to the broader understanding of ferroptosis in eukaryotic microbes.
Collapse
Affiliation(s)
- Min Seok Kwun
- School of Life Science, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Dong Gun Lee
- School of Life Science, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Republic of Korea
- Institute of Life Science and Biotechnology, Kyungpook National University, Daegu 41566, Republic of Korea
| |
Collapse
|
34
|
Wu Q, Zhang X, Qin M, Shi D, Li Y. Dual roles of LncRNA RNA143598: a biomarker for rheumatoid arthritis and its implications in cancer. Clin Rheumatol 2025:10.1007/s10067-025-07448-2. [PMID: 40279008 DOI: 10.1007/s10067-025-07448-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 02/24/2025] [Accepted: 04/15/2025] [Indexed: 04/26/2025]
Abstract
OBJECTIVE Rheumatoid arthritis (RA) is a chronic autoimmune disease with complex pathological mechanisms, including immune system dysregulation and chronic inflammation. Recent studies indicate that long non-coding RNAs (LncRNAs) play key roles in immune regulation and have been implicated in the pathogenesis of multiple diseases, including RA and various types of cancer. Understanding the involvement of LncRNAs in RA and their potential transcriptional effects in cancer could provide novel insights into disease mechanisms and therapeutic targets. METHODS Using the GSE94519 dataset, we analyzed serum LncRNA profiles from RA patients and healthy controls. Differential expression genes (DEGs) were identified using GEO2R, and findings were validated via quantitative polymerase chain reaction (qPCR) in 39 RA and 53 healthy samples. Receiver operating characteristic (ROC) analysis was performed to evaluate diagnostic performance. A pan-cancer analysis of MTRNR2L1 was conducted using TCGA data, with immune infiltration assessed via ssGSEA. RESULTS RNA143598 was significantly upregulated in RA patients, and qPCR confirmed its diagnostic potential (AUC = 0.77). Pan cancer analysis shows that MTRNR2L1 is highly expressed in glioblastoma (GBM) and lowly expressed in head and neck squamous cell carcinoma (HNSC), with high GBM expression linked to poor prognosis. Immune infiltration analysis showed MTRNR2L1 correlated with CD8 + T cells, macrophages, and dendritic cells in GBM. CONCLUSION RNA143598 is a promising RA biomarker, and its transcription gene MTRNR2L1 demonstrates potential in cancer prognosis and immune regulation. These findings provide a foundation for future research on targeted therapies for RA and cancer. Key Points • RNA143598 is identified as a significant biomarker for diagnosing rheumatoid arthritis (RA), showing promise for clinical application. • Quantitative PCR validation demonstrates the diagnostic potential of RNA143598, with an area under the curve (AUC) of 0.77. • MTRNR2L1, which is RNA143598 transcribed gene, exhibits differential expression in different cancer types, with high levels associated with poor prognosis in glioblastoma (GBM). • Immune infiltration analysis links MTRNR2L1 expression to the presence of CD8 + T cells, macrophages, and dendritic cells, suggesting its role in immune regulation in GBM.
Collapse
Affiliation(s)
- Qiuhua Wu
- Department of Clinical Laboratory, First Affiliated Hospital of Huzhou University, The First People's Hospital of Huzhou City, Huzhou, 313000, Zhejiang, China
| | - Xiaoxia Zhang
- Department of Clinical Laboratory, First Affiliated Hospital of Huzhou University, The First People's Hospital of Huzhou City, Huzhou, 313000, Zhejiang, China
| | - Meiyun Qin
- Department of Clinical Laboratory, First Affiliated Hospital of Huzhou University, The First People's Hospital of Huzhou City, Huzhou, 313000, Zhejiang, China
| | - Danfei Shi
- Department of Pathology, First Affiliated Hospital of Huzhou University, The First People's Hospital of Huzhou City, Huzhou, 313000, Zhejiang, China
| | - Yong Li
- Department of Clinical Laboratory, First Affiliated Hospital of Huzhou University, The First People's Hospital of Huzhou City, Huzhou, 313000, Zhejiang, China.
| |
Collapse
|
35
|
Hu R, Tran B, Li S, Stackpole ML, Zeng W, Zhou Y, Melehy A, Sadeghi S, Finn RS, Zhou XJ, Li W, Agopian VG. Noninvasive prognostication of hepatocellular carcinoma based on cell-free DNA methylation. PLoS One 2025; 20:e0321736. [PMID: 40279344 PMCID: PMC12026916 DOI: 10.1371/journal.pone.0321736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 03/11/2025] [Indexed: 04/27/2025] Open
Abstract
BACKGROUND The current noninvasive prognostic evaluation methods for hepatocellular carcinoma (HCC), which are largely reliant on radiographic imaging features and serum biomarkers such as alpha-fetoprotein (AFP), have limited effectiveness in discriminating patient outcomes. Identification of new prognostic biomarkers is a critical unmet need to improve treatment decision-making. Epigenetic changes in cell-free DNA (cfDNA) have shown promise in early cancer diagnosis and prognosis. Thus, we aim to evaluate the potential of cfDNA methylation as a noninvasive predictor for prognostication in patients with active, radiographically viable HCC. METHODS Using Illumina HumanMethylation450 array data of 377 HCC tumors and 50 adjacent normal tissues obtained from The Cancer Genome Atlas (TCGA), we identified 158 HCC-related DNA methylation markers associated with overall survival (OS). This signature was further validated in 29 HCC tumor tissue samples. Subsequently, we applied the signature to an independent cohort of 52 patients with plasma cfDNA samples by calculating the cfDNA methylation-based risk score (methRisk) via random survival forest models with 10-fold cross-validation for the prognostication of OS. RESULTS The cfDNA-based methRisk showed strong discriminatory power when evaluated as a single predictor for OS (3-year AUC = 0.81, 95% CI: 0.68-0.94). Integrating the methRisk with existing risk indices like Barcelona clinic liver cancer (BCLC) staging significantly improved the noninvasive prognostic assessments for OS (3-year AUC = 0.91, 95% CI: 0.80-1), and methRisk remained an independent predictor of survival in the multivariate Cox model (P = 0.007). CONCLUSIONS Our study serves as a pilot study demonstrating that cfDNA methylation biomarkers assessed from a peripheral blood draw can stratify HCC patients into clinically meaningful risk groups. These findings indicate that cfDNA methylation is a promising noninvasive prognostic biomarker for HCC, providing a proof-of-concept for its potential clinical utility and laying the groundwork for broader applications.
Collapse
Affiliation(s)
- Ran Hu
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California, United States of America
- Bioinformatics Interdepartmental Graduate Program, University of California at Los Angeles, Los Angeles, California, United States of America
- Institute for Quantitative and Computational Biosciences, University of California at Los Angeles, Los Angeles, California, United States of America
| | - Benjamin Tran
- Department of Surgery, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California, United States of America
| | - Shuo Li
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California, United States of America
| | - Mary L. Stackpole
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California, United States of America
| | - Weihua Zeng
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California, United States of America
| | - Yonggang Zhou
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California, United States of America
| | - Andrew Melehy
- Department of Surgery, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California, United States of America
| | - Saeed Sadeghi
- Department of Medicine, Division of Hematology/Oncology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California, United States of America
| | - Richard S. Finn
- Department of Medicine, Division of Hematology/Oncology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California, United States of America
| | - Xianghong Jasmine Zhou
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California, United States of America
- Institute for Quantitative and Computational Biosciences, University of California at Los Angeles, Los Angeles, California, United States of America
- Jonsson Comprehensive Cancer Center, University of California at Los Angeles, Los Angeles, California, United States of America
| | - Wenyuan Li
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California, United States of America
| | - Vatche G. Agopian
- Department of Surgery, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California, United States of America
- Jonsson Comprehensive Cancer Center, University of California at Los Angeles, Los Angeles, California, United States of America
| |
Collapse
|
36
|
Kolligundla LP, Sullivan KM, Mukhi D, Andrade-Silva M, Liu H, Guan Y, Gu X, Wu J, Doke T, Hirohama D, Guarnieri P, Hill J, Pullen SS, Kuo J, Inamoto M, Susztak K. Glutathione-specific gamma-glutamylcyclotransferase 1 ( CHAC1) increases kidney disease risk by modulating ferroptosis. Sci Transl Med 2025; 17:eadn3079. [PMID: 40267214 DOI: 10.1126/scitranslmed.adn3079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 08/20/2024] [Accepted: 04/03/2025] [Indexed: 04/25/2025]
Abstract
Genome-wide association studies (GWASs) have identified more than 1000 loci where genetic variants correlate with kidney function. However, the specific genes, cell types, and mechanisms influenced by these genetic variants remain largely uncharted. Here, we identified glutathione-specific gamma-glutamylcyclotransferase 1 (CHAC1) on chromosome 15 as affected by GWAS variants by analyzing human kidney gene expression and methylation information. Both CHAC1 RNA and protein were expressed in the loop of Henle region in mouse and human kidneys, and CHAC1 expression was higher in patients carrying disease risk variants. Using CRISPR technology, we created mice with a single functional copy of the Chac1 gene (Chac1+/-) that displayed no baseline phenotypic alterations in kidney structure or function. These mice demonstrated resilience to kidney disease in multiple models, including folic acid-induced nephropathy, adenine-induced chronic kidney disease, and uninephrectomy-streptozotocin-induced diabetic nephropathy. We further showed that CHAC1 plays a critical role in degrading the cellular antioxidant glutathione. Tubule cells isolated from Chac1+/- mice showed increased glutathione, decreased lipid peroxidation, improved cell viability, and protection against ferroptosis. Expression of ferroptosis-associated genes was also lower in mice with only one copy of Chac1. Higher CHAC1 protein also correlated with ferroptosis-related protein abundance in kidney biopsies from patients with kidney disease. This study positions CHAC1 as an important mediator of kidney disease that influences glutathione concentrations and ferroptosis, suggesting potential avenues to explore for the treatment of kidney diseases.
Collapse
Affiliation(s)
- Lakshmi P Kolligundla
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Penn/CHOP Kidney Innovation Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
| | - Katie M Sullivan
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Department of Pediatrics, Medical College of Wisconsin Pediatric Nephrology, Milwaukee, WI 53226, USA
| | - Dhanunjay Mukhi
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Penn/CHOP Kidney Innovation Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
| | - Magaiver Andrade-Silva
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Penn/CHOP Kidney Innovation Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
| | - Hongbo Liu
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Penn/CHOP Kidney Innovation Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
| | - Yuting Guan
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
| | - Xiangchen Gu
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
| | - Junnan Wu
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
| | - Tomohito Doke
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
| | - Daigoro Hirohama
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Penn/CHOP Kidney Innovation Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
| | - Paolo Guarnieri
- Department of Cardiometabolic Diseases Research, Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT 06877, USA
| | - Jon Hill
- Department of Cardiometabolic Diseases Research, Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT 06877, USA
| | - Steven S Pullen
- Department of Cardiometabolic Diseases Research, Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT 06877, USA
| | - Jay Kuo
- Department of Cardiometabolic Diseases Research, Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT 06877, USA
| | | | - Katalin Susztak
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Penn/CHOP Kidney Innovation Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Department of Genetics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
| |
Collapse
|
37
|
Yan H, Ji X, Li B. Advancing personalized, predictive, and preventive medicine in bladder cancer: a multi-omics and machine learning approach for novel prognostic modeling, immune profiling, and therapeutic target discovery. Front Immunol 2025; 16:1572034. [PMID: 40330458 PMCID: PMC12053186 DOI: 10.3389/fimmu.2025.1572034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Accepted: 03/31/2025] [Indexed: 05/08/2025] Open
Abstract
Objective This study aimed to identify and analyze immunogenic cell death (ICD)-related multi-omics features in bladder cancer (BLCA) using single-cell RNA sequencing (scRNA-seq) and bulk RNA-seq data. By integrating these datasets, we sought to construct a prognostic signature (ICDRS) and explore its clinical and biological implications, including its association with immune cell infiltration, tumor microenvironment (TME), and drug sensitivity. Methods Publicly available datasets from TCGA and GEO, including scRNA-seq (GSE222315, 9 samples) and bulk RNA-seq (TCGA-BLCA, 403 samples; GSE13507, 160 samples), were analyzed. Single-cell data were processed using Seurat, and ICD scores were calculated using single-sample gene set enrichment analysis (ssGSEA). Weighted gene co-expression network analysis (WGCNA) identified ICD-related modules, and machine learning algorithms (Lasso, Ridge, CoxBoost) were employed to construct the ICDRS. Survival analysis, immune infiltration, pathway enrichment, and drug sensitivity were evaluated to validate the model. Results The ICDRS, based on eight key genes (IL32, AHNAK, ANXA5, FN1, GSN, CNN3, FXYD3, CTSS), effectively stratified BLCA patients into high- and low-risk groups with significant differences in overall survival (OS, P < 0.001). High ICDRS scores were associated with immune-suppressive TME, including increased infiltration of T cells CD4 memory resting (P = 0.02) and macrophages M0/M1/M2 (P = 0.01). Pathway enrichment revealed correlations with cholesterol homeostasis, epithelial-mesenchymal transition (EMT), and KRAS signaling. Drug sensitivity analysis showed high-risk groups were resistant to Cisplatin (P = 0.003), Mitomycin C (P = 0.01), and Paclitaxel (P = 0.004), with IC50 values significantly higher than low-risk groups. Conclusion The ICDRS serves as a robust prognostic biomarker for BLCA, offering insights into tumor immune evasion mechanisms and potential therapeutic targets. Its integration with clinical features enhances personalized treatment strategies, highlighting the importance of ICD in BLCA immunotherapy and precision medicine. The model's predictive accuracy and biological relevance were validated across multiple datasets, underscoring its potential for clinical application.
Collapse
Affiliation(s)
- Han Yan
- Department of Pain Medicine, The First Hospital of China Medical University, Liaoning, Shenyang, China
| | - Xinyu Ji
- Department of Thoracic Surgery, The First Hospital of China Medical University, Liaoning, Shenyang, China
| | - Bohan Li
- Department of Urinary Surgery, The First Hospital of China Medical University, Liaoning, Shenyang, China
| |
Collapse
|
38
|
Tang K, Liu M, Zhang C. Construction of a prognostic model and identification of key genes in liver hepatocellular carcinoma based on multi-omics data. Sci Rep 2025; 15:13393. [PMID: 40251374 PMCID: PMC12008308 DOI: 10.1038/s41598-025-98038-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 04/09/2025] [Indexed: 04/20/2025] Open
Abstract
Liver hepatocellular carcinoma (LIHC) strongly contributes to global cancer mortality, highlighting the need for a deeper understanding of its molecular mechanisms to enhance patient prognosis and treatment approaches. We aimed to investigate the differential expression of immunogenic cell death-related genes (ICDRGs) and cellular senescence-related genes (CSRGs) in LIHC and their effects on patient prognosis. We combined the GSE25097, GSE46408, and GSE121248 datasets by eliminating batch effects and standardizing the data. After processing, 16 genes were identified as ICDR&CSR differentially expressed genes (ICDR&CSRDEGs), including UBE2T, HJURP, PTTG1, CENPA, and FOXM1. Gene set enrichment analysis indicated a strong enrichment of these genes in pre-Notch expression and processing. Gene set variation analysis revealed 20 pathways with significant differences between the LIHC and control groups. Mutation analysis identified TP53 as the most commonly mutated gene in LIHC samples. A prognostic risk model integrating 12 ICDR&CSRDEGs was developed, showing high precision at 1 year but diminished accuracy at 2 and 3 years. Our constructed prognostic risk model provides valuable insights for predicting patient outcomes and may guide future therapeutic interventions targeting these specific genes. Further research is needed to explore the mechanistic roles of these genes in LIHC progression and treatment response.
Collapse
Affiliation(s)
- Kun Tang
- Department of Hepatobiliary Surgery, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong Province, China
| | - Mingjiang Liu
- Department of Hepatobiliary Surgery, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong Province, China
| | - Cuisheng Zhang
- Department of Hepatobiliary Surgery, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong Province, China.
| |
Collapse
|
39
|
Gong W, Wang Y, Li Q, Gao Y, Li J. Regulation of SLC7A11 by LncRNA GPRC5D-AS1 mediates ferroptosis in skeletal muscle: Mechanistic exploration of sarcopenia. Front Mol Biosci 2025; 12:1557218. [PMID: 40309008 PMCID: PMC12040812 DOI: 10.3389/fmolb.2025.1557218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 04/04/2025] [Indexed: 05/02/2025] Open
Abstract
Sarcopenia is a chronic, progressive disease characterized by the gradual loss of skeletal muscle strength and mass. This study investigates the role of the long non-coding RNA GPRC5D-AS1 in the development and progression of sarcopenia through its regulation of SLC7A11. Skeletal muscle samples were obtained from sarcopenia patients and healthy controls to assess the expression levels of GPRC5D-AS1 and SLC7A11. Flow cytometry was used to evaluate iron content, lipid peroxidation, and antioxidant markers. A ferroptosis model was established in human skeletal muscle cells (HSKM) using the inducer erastin, and GPRC5D-AS1 overexpression plasmids were introduced to observe their effects on cell proliferation and ferroptosis indicators. In the sarcopenia group, both GPRC5D-AS1 and SLC7A11 expression levels decreased significantly, along with SLC7A11 protein translation. Erastin treatment markedly reduced cell viability and increased iron content, elevating ferroptosis marker genes (COX2, ACSL4, PTGS2, NOX1) while reducing GPX4 and FTH1 levels. The overexpression of GPRC5D-AS1 reversed these changes, enhancing antioxidant capacity and cell survival. Conversely, silencing SLC7A11 diminished the protective effects of GPRC5D-AS1 on cell proliferation and ferroptosis. These findings suggest that GPRC5D-AS1 overexpression increases SLC7A11 expression and reduces ferroptosis incidence in HSKM.
Collapse
Affiliation(s)
- Wei Gong
- Department of Critical Care Medicine, The First Hospital of Jilin University, Changchun, China
| | - Yan Wang
- Department of Geriatrics and Special medical treatment, The First Hospital of Jilin University, Changchun, China
| | - Qun Li
- Health Examination Center, The First Hospital of Jilin University, Changchun, China
| | - Yating Gao
- Department of Geriatrics and Special medical treatment, The First Hospital of Jilin University, Changchun, China
| | - Jie Li
- Department of Geriatrics and Special medical treatment, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
40
|
Ji PX, Zhang P, Zhou HL, Yu H, Fu Y. MEX3A promotes cell proliferation by regulating the RORA/β-catenin pathway in hepatocellular carcinoma. World J Gastrointest Oncol 2025; 17:102084. [PMID: 40235874 PMCID: PMC11995337 DOI: 10.4251/wjgo.v17.i4.102084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 01/27/2025] [Accepted: 02/14/2025] [Indexed: 03/25/2025] Open
Abstract
BACKGROUND MEX3A is a member of the human homologous gene MEX-3 family. It has been shown to promote cell proliferation and migration in various cancers, indicating its potential clinical significance. However, the role of MEX3A in hepatocellular carcinoma (HCC) remains largely unexplored, with limited reports available in the literature. AIM To investigate expression and clinical significance of MEX3A in HCC and explore its potential role in tumor progression. METHODS We analyzed MEX3A mRNA expression in HCC and adjacent tissues using data from The Cancer Genome Atlas (TCGA). The correlation between MEX3A expression and overall survival (OS) was evaluated. Immunohistochemistry was performed on HCC surgical specimens to validate MEX3A expression and its association with clinical parameters, including hepatitis B virus (HBV) positivity, tumor differentiation and tumor size. Additionally, MEX3A knockdown HCC cell lines were constructed to explore the biological functions of MEX3A. Cell proliferation was assessed using cell counting kit-8 and clone formation assays, while cell cycle progression was analyzed by flow cytometry. The effects of MEX3A on the Wnt/β-catenin signaling pathway were examined by western blotting and immunofluorescence. Cell migration was evaluated using scratch and Transwell assays. Finally, the role of the transcription factor RORA in mediating MEX3A effects was explored by silencing RORA and analyzing its impact on cell proliferation and protein expression. RESULTS TCGA data analysis revealed that MEX3A mRNA expression was significantly higher in HCC tissues compared to adjacent tissues. Higher MEX3A expression was associated with poorer OS. These findings were validated in HCC surgical specimens. Immunohistochemistry confirmed elevated MEX3A expression in HCC tissues and showed positive correlations with Ki-67 and vimentin levels. MEX3A expression was closely related to HBV positivity, tumor differentiation and tumor size. Mechanistic studies demonstrated that MEX3A knockdown inhibited cell proliferation and cell cycle progression, as shown by reduced expression of β-catenin, c-Myc and cyclin D1. Additionally, MEX3A knockdown inhibited the nuclear entry of β-catenin, thereby suppressing the activation of downstream oncogenic pathways. MEX3A depletion significantly reduced the migratory ability of HCC cells, likely through downregulation of the epithelial-mesenchymal transition pathway. Transcription factor analysis identified RORA as a potential mediator of MEX3A effects. Silencing RORA antagonized the effects of MEX3A on cell proliferation and the expression of β-catenin, c-Myc and cyclin D1. CONCLUSION MEX3A promotes cell proliferation in HCC by regulating the RORA/β-catenin pathway. Our findings suggest that MEX3A could serve as a prognostic marker and therapeutic target for HCC.
Collapse
Affiliation(s)
- Peng-Xiang Ji
- Hand Surgery Laboratory, Suzhou Ruihua Orthopedic Hospital, Suzhou Medical College of Soochow University, Suzhou 215104, Jiangsu Province, China
| | - Ping Zhang
- Hand Surgery Laboratory, Suzhou Ruihua Orthopedic Hospital, Suzhou Medical College of Soochow University, Suzhou 215104, Jiangsu Province, China
| | - Hui-Ling Zhou
- Department of Pathology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou 225300, Jiangsu Province, China
| | - Hong Yu
- Department of Pathology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou 225300, Jiangsu Province, China
| | - Yi Fu
- Department of Human Anatomy, Histology and Embryology, Suzhou Medical College of Soochow University, Suzhou 215123, Jiangsu Province, China
| |
Collapse
|
41
|
Yang Q, Zhang H, Luo J, Yu H, Yang X, Wang C. FADS2 inhibits colorectal cancer cell proliferation by regulating ferroptosis through SLC7A11/GPX4. Mol Biol Rep 2025; 52:394. [PMID: 40232565 DOI: 10.1007/s11033-025-10395-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 02/26/2025] [Indexed: 04/16/2025]
Abstract
BACKGROUND Colorectal cancer (CRC) is a leading factor in cancer mortality globally. Ferroptosis, a regulated cell death described via lipid peroxidation, is crucial in cancer biology. This study explores the link between ferroptosis, FADS2, and CRC, focusing on the prognostic significance and therapeutic potential of targeting FADS2. METHODS The differential expression analysis of the Cancer Genome Atlas-colon adenocarcinoma (TCGA-COAD) and GSE36400 datasets was conducted to determine key ferroptosis-related genes, followed by functional enrichment analysis. Prognosis-related genes were assessed utilizing Least Absolute Shrinkage and Selection Operator (LASSO) Cox regression. Genetic variation analysis and immune analysis were employed to evaluate the clinical significance of FADS2. The impacts of FADS2 knockdown on CRC cell migration, proliferation, invasion, and ferroptosis were evaluated by in vitro cell experiments. RESULTS 64 key ferroptosis-related genes in CRC were highly enriched in pathways such as glutathione metabolism and peroxisome. Eleven prognosis-associated genes were identified, with TP53 showing the highest mutation frequency. High FADS2 expression was linked to poorer prognosis and higher immune cell infiltration. FADS2 knockdown significantly decreased glutathione (GSH) levels, SLC7A11, and GPX4 expression, increased malondialdehyde (MDA) levels, indicating the promotion of ferroptosis. Functional tests revealed knockdown FADS2 repressed CRC cell proliferation, migration, and invasion. SLC7A11 or GPX4 overexpression partially rescued the effects of FADS2 knockdown. Additionally, FADS2 knockdown enhances the chemosensitivity of CRC cells to oxaliplatin. CONCLUSION FADS2 is essential for encouraging CRC cell proliferation and tumor growth by preventing ferroptosis. Targeting FADS2 may enhance ferroptosis and suppress CRC progression, offering a possible course of treatment for CRC patients. The knockdown of FADS2 enhances the chemosensitivity of CRC cells to oxaliplatin, providing valuable insights for future clinical applications.
Collapse
Affiliation(s)
- Qinghui Yang
- Department of Oncology, Minhang Branch, Zhongshan Hospital, Fudan University, No. 170 Xinsong Road, Minhang District, Shanghai, China
- Key Laboratory of Whole-Period Monitoring and Precise Intervention of Digestive Cancer(SMHC), Minhang Hospital & AHS, Fudan University, Shanghai, China
| | - Hao Zhang
- Department of Oncology, Minhang Branch, Zhongshan Hospital, Fudan University, No. 170 Xinsong Road, Minhang District, Shanghai, China
- Key Laboratory of Whole-Period Monitoring and Precise Intervention of Digestive Cancer(SMHC), Minhang Hospital & AHS, Fudan University, Shanghai, China
| | - Jing Luo
- Department of Oncology, Minhang Branch, Zhongshan Hospital, Fudan University, No. 170 Xinsong Road, Minhang District, Shanghai, China
- Key Laboratory of Whole-Period Monitoring and Precise Intervention of Digestive Cancer(SMHC), Minhang Hospital & AHS, Fudan University, Shanghai, China
| | - Hongmei Yu
- Department of Oncology, Minhang Branch, Zhongshan Hospital, Fudan University, No. 170 Xinsong Road, Minhang District, Shanghai, China
- Key Laboratory of Whole-Period Monitoring and Precise Intervention of Digestive Cancer(SMHC), Minhang Hospital & AHS, Fudan University, Shanghai, China
| | - Xiaodi Yang
- Department of Oncology, Minhang Branch, Zhongshan Hospital, Fudan University, No. 170 Xinsong Road, Minhang District, Shanghai, China.
- Key Laboratory of Whole-Period Monitoring and Precise Intervention of Digestive Cancer(SMHC), Minhang Hospital & AHS, Fudan University, Shanghai, China.
| | - Chen Wang
- Department of Oncology, Minhang Branch, Zhongshan Hospital, Fudan University, No. 170 Xinsong Road, Minhang District, Shanghai, China.
- Key Laboratory of Whole-Period Monitoring and Precise Intervention of Digestive Cancer(SMHC), Minhang Hospital & AHS, Fudan University, Shanghai, China.
| |
Collapse
|
42
|
Ababneh E, Velez S, Zhao J. Immune evasion and resistance in breast cancer. Am J Cancer Res 2025; 15:1517-1539. [PMID: 40371160 PMCID: PMC12070088 DOI: 10.62347/pngt6996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 12/18/2024] [Indexed: 05/16/2025] Open
Abstract
Breast cancer (BC) is the most common malignancy in females with an increasing incidence in the last decade. The previously observed decline in BC mortality rates has also slowed down recently with an increase in the incidence of invasive BC. BC has various molecular subtypes. Among these subtypes, triple-negative breast cancer (TNBC) represents the most aggressive BC, with a poor prognosis. Because lack of the hormonal or human epidermal growth factor receptor 2 (HER2) receptors, TNBC is resistant to hormonal and HER2 targeted therapy effective for other BC subtypes. The good news is that TNBC has recently been considered an immunologically 'hot' tumor. Therefore, immunotherapy, particularly immune checkpoint inhibitor therapy, represents a promising therapeutic approach TNBC. However, a considerable percentage of patients with TNBC do not respond well to immunotherapy, indicating that TNBC seems to adopt several mechanisms to evade immune surveillance. Thus, it is crucial to investigate the mechanisms underlying TNBC immune evasion and resistance to immunotherapy. In this review, we examine and discuss the most recently discovered mechanisms for BC, with a particular focus on TNBC, to evade the immune surveillance via kidnapping the immune checkpoints, suppressing the immune responses in tumor microenvironment and inhibiting the tumor antigen presentation. Evaluation of these mechanisms in BC will hopefully guide future immunotherapeutic research and clinical trials.
Collapse
Affiliation(s)
- Ebaa Ababneh
- Burnett School of Biomedical Science, Medical College, University of Central Florida Orlando, FL, USA
| | - Sarah Velez
- Burnett School of Biomedical Science, Medical College, University of Central Florida Orlando, FL, USA
| | - Jihe Zhao
- Burnett School of Biomedical Science, Medical College, University of Central Florida Orlando, FL, USA
| |
Collapse
|
43
|
Dong H, Zhang Z, Fu C, Guo M, Zhang H, Cai X, Cheng H. Association between fibrosis-4 index (FIB-4) and gallstones: an analysis of the NHANES 2017-2020 cross-sectional study. BMC Gastroenterol 2025; 25:229. [PMID: 40197261 PMCID: PMC11977908 DOI: 10.1186/s12876-025-03809-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Accepted: 03/20/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Gallstones are a common digestive disorder, yet the association between the fibrosis-4 index (FIB-4) and gallstone formation remains poorly understood. This study explores the link between FIB-4 levels and gallstone prevalence among US adults. METHODS This study was based on data from the 2017-2020 National Health and Nutrition Examination Survey (NHANES), which included 7,771 participants. The association between FIB-4 and gallstone risk was analyzed using multivariate logistic regression with restricted cubic spline (RCS) analysis to assess nonlinear correlations, and threshold effects analysis to identify inflection points. Robustness checks included subgroup analysis. RESULTS The weighted prevalence of gallstones in this study was 11%. Multiple logistic regression analysis showed that FIB-4 levels were significantly and positively associated with the risk of gallstones. In the fully adjusted model, each unit increase in FIB-4 as a continuous variable increased the risk of gallstones by 19% (OR = 1.19, 95% CI: 1.10, 1.29). When FIB-4 was grouped by quartiles, the risk of gallstones was increased by 60% (OR = 1.60, 95% CI: 1.25, 2.03) in the Q4 group compared to the Q1 group. RCS analysis further revealed a nonlinear positive correlation between FIB-4 and gallstones (P for nonlinear = 0.015) with an inflection point at 2.43, (P for log likelihood ratio test = 0.001). Bonferroni-corrected subgroup analyses showed that the association of FIB-4 with gallstones was statistically significant among non-Hispanic whites, those without heart failure, those without coronary heart disease, alcohol drinkers, and smokers (P < 0.00217). CONCLUSION In this study, we found that elevated levels of FIB-4 were significantly and positively association with the risk of gallstones, and showed a non-linear trend. FIB-4 may have a potential application in the risk assessment of gallstones.
Collapse
Affiliation(s)
- Huqiang Dong
- School of Public Health, Ningxia Medical University, Yinchuan, Ningxia, 750004, China
| | - Zufa Zhang
- Department of Urology, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, 116001, China
| | - Chang Fu
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Mixue Guo
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, Ningxia, 750004, China
| | - Haifeng Zhang
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Xintian Cai
- Heart, Lung and Vessels Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610072, China.
| | - Hongping Cheng
- Center of Health Administration and Development Studies, Hubei University of Medicine, Shiyan, Hubei, 442000, China.
| |
Collapse
|
44
|
Ma Z, Zheng M, Liu P. Identification of fatty acid metabolism-related genes in the tumor microenvironment of breast cancer by a development and validation of prognostic index signature. Hereditas 2025; 162:55. [PMID: 40197314 PMCID: PMC11974137 DOI: 10.1186/s41065-025-00425-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Accepted: 03/26/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Breast cancer (BRCA) is a malignancy originating in the breast cells, characterized by a poor overall survival rate. Post-resection, chemotherapy is commonly recommended as a primary therapeutic approach; however, its efficacy remains limited. Recent advancements in lipidomics and metabolomics have provided new insights into the intricate landscape of fatty acid metabolism (FAM) and the fatty acid lipidome in both health and disease. A growing body of evidence suggests that dysregulations in FAM and fatty acid levels play a significant role in cancer initiation and progression. Despite these advances, the precise mechanisms through which FAM mediates the anti-cancer effects of lobaplatin in BRCA remain poorly understood and warrant further investigation. METHODS GEO and TCGA data were classified into two types. We aimed to show how FAMGs influence immune function, immune checkpoints, and m6a in BRCA. A co-expression analysis discovered that gene expression is strongly connected to pyroptosis. The TCGA gathered information about mRNAsi, gene mutations, CNV, and clinical features. RESULTS In the low-risk group, overall survival (OS) is longer. GSEA was utilized to identify immune and tumor-related pathways. Most of the FAMG-derived prognostic signatures predominantly modulate immunological and oncogenic signaling pathways, including the Wnt, neurotrophin, chemokine, and calcium signaling cascades. Among the genes involved are CEL, WT1, and ULBP2. Expression levels varied as well. The prognostic model, CNVs, single nucleotide polymorphism (SNP), and drug sensitivity all pointed to the gene. CONCLUSIONS The primary objective of this study is to identify and validate BRCA-associated FAMGs that can serve as prognostic indicators and provide insights into immune system function, while also offering evidence to support the development of fatty acid metabolism-related molecularly targeted therapeutics. Consequently, FAMGs and their interactions with the immune system, as well as their role in BRCA, may emerge as promising therapeutic targets.
Collapse
Affiliation(s)
- Zhaofeng Ma
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, China
| | - Man Zheng
- Dongying People's Hospital (Dongying Hospital of Shandong Provincial Hospital Group), Dongying, Shandong Province, 257091, China
| | - Pulin Liu
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, China.
| |
Collapse
|
45
|
de Alwis N, Beard S, Baird L, Binder NK, Pritchard N, Tong S, Kaitu'u-Lino TJ, Hui L, Hannan NJ. Phosphoglucomutase 5 gene transcripts are expressed by the human placenta and differentially regulated in placental dysfunction. Sci Rep 2025; 15:11381. [PMID: 40180976 PMCID: PMC11968825 DOI: 10.1038/s41598-025-94498-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 03/14/2025] [Indexed: 04/05/2025] Open
Abstract
The placenta plays an essential role facilitating nutrient, gas and waste exchange between the maternal and fetal systems for optimal fetal growth. When placental development is impaired and the placenta dysfunctional, serious pregnancy complications such as fetal growth restriction and preeclampsia may arise. Previously, phosphoglucomutase-5 (PGM5) transcripts were found to be highly elevated in the blood of patients whose pregnancies were complicated by fetal growth restriction and preeclampsia. As both conditions feature placental insufficiency, here we aimed to characterise PGM5 levels in the healthy and dysfunctional placenta. PGM5 expression was detectable in all placental samples across gestation, in cases of preterm preeclampsia, fetal growth restriction and controls. PGM5 mRNA expression was significantly downregulated in the pathological placentas compared to controls, but PGM5 protein production was not dysregulated. Isolated cytotrophoblast and placental explant tissue exposed to hypoxia (modelling placental dysfunction) demonstrated significantly increased PGM5 expression, but again did not change protein levels. Silencing PGM5 expression under hypoxic conditions in primary cytotrophoblast did not alter anti-angiogenic sFLT-1 secretion but increased expression of multiple genes associated with cell growth, apoptosis and oxidative stress, whilst also increasing cell viability. Expression of PGM5 in all placental samples assessed suggests that PGM5 has functions in the placenta. However, further investigation could be performed to explore the discrepancies in protein and mRNA expression, as well as the precise function of PGM5 in the placenta, and whether altered PGM5 levels may be important for placental development.
Collapse
Affiliation(s)
- Natasha de Alwis
- Department of Obstetrics, Gynaecology and Newborn Health, The University of Melbourne and Mercy Hospital for Women, 163 Studley Rd, Heidelberg, Melbourne, VIC, 3084, Australia
- Northern Health, Epping, Melbourne, VIC, 3076, Australia
| | - Sally Beard
- Department of Obstetrics, Gynaecology and Newborn Health, The University of Melbourne and Mercy Hospital for Women, 163 Studley Rd, Heidelberg, Melbourne, VIC, 3084, Australia
- Northern Health, Epping, Melbourne, VIC, 3076, Australia
| | - Lydia Baird
- Department of Obstetrics, Gynaecology and Newborn Health, The University of Melbourne and Mercy Hospital for Women, 163 Studley Rd, Heidelberg, Melbourne, VIC, 3084, Australia
- Northern Health, Epping, Melbourne, VIC, 3076, Australia
| | - Natalie K Binder
- Department of Obstetrics, Gynaecology and Newborn Health, The University of Melbourne and Mercy Hospital for Women, 163 Studley Rd, Heidelberg, Melbourne, VIC, 3084, Australia
| | - Natasha Pritchard
- Department of Obstetrics, Gynaecology and Newborn Health, The University of Melbourne and Mercy Hospital for Women, 163 Studley Rd, Heidelberg, Melbourne, VIC, 3084, Australia
| | - Stephen Tong
- Department of Obstetrics, Gynaecology and Newborn Health, The University of Melbourne and Mercy Hospital for Women, 163 Studley Rd, Heidelberg, Melbourne, VIC, 3084, Australia
| | - Tu'uhevaha J Kaitu'u-Lino
- Department of Obstetrics, Gynaecology and Newborn Health, The University of Melbourne and Mercy Hospital for Women, 163 Studley Rd, Heidelberg, Melbourne, VIC, 3084, Australia
| | - Lisa Hui
- Department of Obstetrics, Gynaecology and Newborn Health, The University of Melbourne and Mercy Hospital for Women, 163 Studley Rd, Heidelberg, Melbourne, VIC, 3084, Australia
- Northern Health, Epping, Melbourne, VIC, 3076, Australia
| | - Natalie J Hannan
- Department of Obstetrics, Gynaecology and Newborn Health, The University of Melbourne and Mercy Hospital for Women, 163 Studley Rd, Heidelberg, Melbourne, VIC, 3084, Australia.
- Northern Health, Epping, Melbourne, VIC, 3076, Australia.
| |
Collapse
|
46
|
Song H, Gao M, Bao Z, Yang Y, Chen Y, Hao J. Expression and clinical significance of ITGA3 in breast cancer. Open Med (Wars) 2025; 20:20241113. [PMID: 40181838 PMCID: PMC11967466 DOI: 10.1515/med-2024-1113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 11/11/2024] [Accepted: 11/19/2024] [Indexed: 04/05/2025] Open
Abstract
Background Integrin subunit Alpha 3 is essential for cell adhesion and movement, but its role in breast cancer (BC) is unclear. This study evaluated ITGA3 expression in BC and its clinical significance. Methods The Human Protein Atlas (HPA), UALCAN, and Kaplan-Meier plotter were used to analyze ITGA3 in BC. ITGA3 was evaluated using receiver operating characteristic curves. The cell counting kit-8 test was used to examine the role of ITGA3 in cell proliferation. Results BC tissues' ITGA3 protein and mRNA levels were significantly lower than normal controls. ITGA3 was associated with better recurrence-free survival (RFS) and distant metastasis-free survival, especially in estrogen receptor (ER)-positive, Luminal B, and Luminal B subtypes. ITGA3 predicted RFS for 5 years and the response to chemotherapy. ITGA3 was associated with ER status but not age, tumor, node, metastasis stages, or tumor size. ITGA3 has a positive impact on BC cell growth. Conclusions ITGA3 may be a predictive marker for BC and a therapeutic target. These findings need to be confirmed, and the molecular mechanisms of ITGA3 need to be clarified.
Collapse
Affiliation(s)
- Huachun Song
- Department of Thyroid Breast Oncology, Yiwu Central Hospital, Yiwu, 322099, Zhejiang, China
| | - Meihui Gao
- Clinical Oncology College, Fujian Medical University, Fuzhou, 350122, Fujian, China
| | - Zheng Bao
- Operating Room, Yiwu Central Hospital, Yiwu, 322099, Zhejiang, China
| | - Yong Yang
- Department of Pathology, Yiwu Central Hospital, Yiwu, 322099, Zhejiang, China
| | - Yang Chen
- Department of Thyroid Breast Oncology, Yiwu Central Hospital, Yiwu, 322099, Zhejiang, China
| | - Jing Hao
- Department of Thyroid Breast Oncology, Yiwu Central Hospital, No. 519, Nanmen Street, Yiwu, 322099, Zhejiang, China
| |
Collapse
|
47
|
Tabatabaeian Nimavard R, Sadeghi SA, Mahmoudi M, Zhu G, Sun L. Top-Down Proteomic Profiling of Protein Corona by High-Throughput Capillary Isoelectric Focusing-Mass Spectrometry. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2025; 36:778-786. [PMID: 40025702 PMCID: PMC11964827 DOI: 10.1021/jasms.4c00463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 01/06/2025] [Accepted: 02/19/2025] [Indexed: 03/04/2025]
Abstract
In the rapidly evolving field of nanomedicine, understanding the interactions between nanoparticles (NPs) and biological systems is crucial. A pivotal aspect of these interactions is the formation of a protein corona when NPs are exposed to biological fluids (e.g., human plasma), which significantly influences their behavior and functionality. This study introduces an advanced capillary isoelectric focusing tandem mass spectrometry (cIEF-MS/MS) platform designed to enable high-throughput and reproducible top-down proteomic analysis of protein corona. Our cIEF-MS/MS technique completed each analysis within 30 min. It produced reproducible proteoform measurements of protein corona for at least 50 runs regarding the proteoforms' migration time [relative standard deviations (RSDs) <4%], the proteoforms' intensity (Pearson's correlation coefficients between any two runs >0.90), the number of proteoform identifications (71 ± 10), and the number of proteoform-spectrum matches (PrSMs) (196 ± 30). Of the 53 identified genes, 33 are potential biomarkers of various diseases (e.g., cancer, cardiovascular disease, and Alzheimer's disease). We identified 1-102 proteoforms per potential protein biomarker, containing various sequence variations or post-translational modifications. Delineating proteoforms in protein corona by our cIEF-MS/MS in a reproducible and high-throughput fashion will benefit our understanding of nanobiointeractions and advance both diagnostic and therapeutic nanomedicine technologies.
Collapse
Affiliation(s)
| | - Seyed Amirhossein Sadeghi
- Department
of Chemistry, Michigan State University, 578 S Shaw Lane, East Lansing, Michigan 48824, United States
| | - Morteza Mahmoudi
- Precision
Health Program, Michigan State University, East Lansing, Michigan 48824, United States
- Department
of Radiology, College of Human Medicine, Michigan State University, East
Lansing, Michigan 48824, United States
| | - Guijie Zhu
- Department
of Chemistry, Michigan State University, 578 S Shaw Lane, East Lansing, Michigan 48824, United States
| | - Liangliang Sun
- Department
of Chemistry, Michigan State University, 578 S Shaw Lane, East Lansing, Michigan 48824, United States
| |
Collapse
|
48
|
Tao Q, Li Y, Zhang W, Zhang M, Li X, Jin H, Zheng J, Li Y. Long non-coding RNA ZFAS1 promotes ferroptosis by regulating the miR-185-5p/SLC25A28 axis in clear cell renal cell carcinoma. Int J Biol Macromol 2025; 304:140602. [PMID: 39922352 DOI: 10.1016/j.ijbiomac.2025.140602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 08/04/2024] [Accepted: 11/11/2024] [Indexed: 02/10/2025]
Abstract
Ferroptosis is a novel, iron-dependent regulated cell death mode. The biochemical features of ferroptosis include iron accumulation, lipid peroxidation, inhibition of glutathione peroxidase 4 (GPX4) and antioxidant glutathione (GSH) decrease through inhibition of the system xc- transporter. Zinc finger NFX1 type-containing 1 (ZNFX1) antisense RNA 1 (ZFAS1) is a long non-coding RNA that has been identified as an oncogene in various types of cancers. However, its regulatory role and molecular mechanisms in clear cell renal cell carcinoma (ccRCC) ferroptosis remain unclear. In this study, the ferroptosis inducers (FINS) (erastin and RSL3) were found to increase ZFAS1 expression through the facilitation of SP1 binding to the ZFAS1 promoter. ZFAS1 increased mRNA and protein levels of solute carrier family 25 member 28 (SLC25A28) via functioning as a miR-185-5p sponge. Overexpressed SLC25A28 increased the production of ROS and caused a decrease in NADPH and GSH in cells treated with FINS. In addition, overexpression of ZFAS1 enhanced ferroptosis both in vitro and in vivo. Altogether, this study demonstrates that ZFAS1 is a crucial element of ferroptosis in ccRCC, as it is responsible for the regulation of miR-185-5p and SLC25A28. Introducing ferroptosis could be a beneficial approach to treat ccRCC patients with high ZFAS1 levels.
Collapse
Affiliation(s)
- Qiqi Tao
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yifei Li
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Weizhi Zhang
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Minghong Zhang
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xinmiao Li
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Hui Jin
- Department of Pharmacy, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jianjian Zheng
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
| | - Yeping Li
- Department of Urology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China.
| |
Collapse
|
49
|
Cui Y, Wu A, Liu H, Zhong Y, Yi K. The effect and potential mechanisms of per- and polyfluoroalkyl substances (PFAS) exposure on kidney stone risk. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 294:118087. [PMID: 40157329 DOI: 10.1016/j.ecoenv.2025.118087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 03/16/2025] [Accepted: 03/18/2025] [Indexed: 04/01/2025]
Abstract
Exposure to per- and polyfluoroalkyl substances (PFAS) may be associated with an increased prevalence of some kidney diseases. Kidney stones are common and have a high prevalence of kidney diseases. However, there is no evidence for the effect and potential mechanisms of PFAS on kidney stone risk. In this study, we designed a cross-sectional study using the National Health and Nutrition Examination Surveys (NHANES) data from 2017 to 2020. Our results revealed that PFAS were positively associated with kidney stone risk, and PFDA was the main contributing compound among PFAS. The triglyceride-glucose (TyG) index and the systemic immune-inflammatory (SII) index had significant mediation effects. In addition, target proteins, such as IL-6, TNF, ALB, IL-1B, and AKT1, and signaling pathways, including TNF and IL-17 pathways, might be potential mechanisms of PFAS in promoting kidney stone risk. In conclusion, PFAS, especially PFDA, increases the risk of kidney stones by the mediation effects of the TyG index and SII index. TNF and IL-17 signaling pathways may be potential mechanisms. Our findings provide new evidence for the effects and potential mechanisms of PFAS exposure in increasing kidney stone risk. However, in the future, it is still imperative to further explore and validate the underlying mechanisms of PFAS-induced kidney stone formation through experimental studies.
Collapse
Affiliation(s)
- Ying Cui
- Guangdong Food and Drug Vocational College, Guangzhou 510520, China
| | - Aitong Wu
- China Agriculture University, Beijing 100083, China
| | - Hao Liu
- Guangdong Food and Drug Vocational College, Guangzhou 510520, China
| | - Yuanyuan Zhong
- Department of Pharmacy, The Third People's Hospital of Yunnan Province, Kunming, Yunnan 650011, China
| | - Kefan Yi
- Clinical Nutrition Department, Shanghai Deji Hospital, Qingdao University, Shanghai 200331, China.
| |
Collapse
|
50
|
Duan H, Gao L, Asikaer A, Liu L, Huang K, Shen Y. Prognostic Model Construction of Disulfidptosis-Related Genes and Targeted Anticancer Drug Research in Pancreatic Cancer. Mol Biotechnol 2025; 67:1463-1482. [PMID: 38575817 DOI: 10.1007/s12033-024-01131-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 02/19/2024] [Indexed: 04/06/2024]
Abstract
Pancreatic cancer stands as one of the most lethal malignancies, characterized by delayed diagnosis, high mortality rates, limited treatment efficacy, and poor prognosis. Disulfidptosis, a recently unveiled modality of cell demise induced by disulfide stress, has emerged as a critical player intricately associated with the onset and progression of various cancer types. It has emerged as a promising candidate biomarker for cancer diagnosis, prognosis assessment, and treatment strategies. In this study, we have effectively established a prognostic risk model for pancreatic cancer by incorporating multiple differentially expressed long non-coding RNAs (DElncRNAs) closely linked to disulfide-driven cell death. Our investigation delved into the nuanced relationship between the DElncRNA-based predictive model for disulfide-driven cell death and the therapeutic responses to anticancer agents. Our findings illuminate that the high-risk subgroup exhibits heightened susceptibility to the small molecule compound AZD1208, positioning it as a prospective therapeutic agent for pancreatic cancer. Finally, we have elucidated the underlying mechanistic potential of AZD1208 in ameliorating pancreatic cancer through its targeted inhibition of the peroxisome proliferator-activated receptor-γ (PPARG) protein, employing an array of comprehensive analytical methods, including molecular docking and molecular dynamics (MD) simulations. This study explores disulfidptosis-related genes, paving the way for the development of targeted therapies for pancreatic cancer and emphasizing their significance in the field of oncology. Furthermore, through computational biology approaches, the drug AZD1208 was identified as a potential treatment targeting the PPARG protein for pancreatic cancer. This discovery opens new avenues for exploring targets and screening drugs for pancreatic cancer.
Collapse
Affiliation(s)
- Hongtao Duan
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 405400, People's Republic of China
| | - Li Gao
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 405400, People's Republic of China
| | - Aiminuer Asikaer
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 405400, People's Republic of China
| | - Lingzhi Liu
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 405400, People's Republic of China
| | - Kuilong Huang
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 405400, People's Republic of China
| | - Yan Shen
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 405400, People's Republic of China.
| |
Collapse
|